1
|
Yu S, Wei X, Long F, Gu H, Hao Z. Quantitative Proteomic Analysis Indicates That Pggt1b Deficiency Promotes Cytokine Secretion in Resiquimod-Stimulated Bone Marrow-Derived Macrophages via the NF-κB Pathway. Immun Inflamm Dis 2025; 13:e70185. [PMID: 40192076 PMCID: PMC11973730 DOI: 10.1002/iid3.70185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 03/13/2025] [Accepted: 03/20/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Psoriasis is a systemic inflammatory skin disease mediated by the innate and adaptive immune systems. Recent studies have indicated that macrophages may contribute to the pathogenesis of psoriasis. However, the role of macrophage protein geranylgeranyl transferase type-1β subunit (PGGT1B) in psoriasis is unclear. In this study, we aimed to establish how a reduction in Pggt1b expression in monocytes influences the onset and progression of psoriasis. METHODS Myeloid cell-specific Pggt1b knockout mice were generated, and their bone marrow-derived macrophages (BMDMs) were stimulated with resiquimod (R848) to mimic the psoriatic immune microenvironment. The proteomic analysis enabled us to identify 17 differentially expressed proteins associated with Pggt1b deficiency in the psoriasis macrophage model (folded change ≥ 1.3 and p < 0.05). Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment was performed. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and western blot assays were used to verify the differentially expressed proteins and signaling pathways. Finally, an enzyme-linked immunosorbent assay was used to verify the expression of the key inflammatory cytokine interleukin (IL)-1β. RESULTS In total, six proteins (Dlgap5, Fas, Fnta, Nlrp3, Cd14, and Ticam2) were identified as hub proteins. Furthermore, we found that Pggt1b might mediate R848-induced inflammation via the small G-proteins Rac1 or Cdc42. We found that Pggt1b positively regulates pro-inflammatory responses in R848-stimulated BMDMs via the NF-κB signaling pathway. CONCLUSIONS This study clarified that PGGT1B affected the synthesis of inflammatory cytokines via NF-κB pathway and provided insights into the mechanisms underlying immune responses and inflammation.
Collapse
Affiliation(s)
- Shanshan Yu
- Institute of DermatologyChinese Academy of Medical Sciences & Peking Union Medical CollegeNanjingChina
| | - Xuecui Wei
- School of Public HealthNanjing Medical UniversityNanjingChina
| | - Fangyuan Long
- Institute of DermatologyChinese Academy of Medical Sciences & Peking Union Medical CollegeNanjingChina
| | - Heng Gu
- Institute of DermatologyChinese Academy of Medical Sciences & Peking Union Medical CollegeNanjingChina
| | - Zhimin Hao
- Institute of DermatologyChinese Academy of Medical Sciences & Peking Union Medical CollegeNanjingChina
| |
Collapse
|
2
|
Palazzo E, Lotti R, Quadri M, Pincelli C, Marconi A. IL-17 Ligand and Receptor Family Members Are Differentially Expressed by Keratinocyte Subpopulations and Modulate Their Differentiation and Inflammatory Phenotype. Int J Mol Sci 2025; 26:2989. [PMID: 40243580 PMCID: PMC11988555 DOI: 10.3390/ijms26072989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/21/2025] [Accepted: 03/22/2025] [Indexed: 04/18/2025] Open
Abstract
Psoriasis is a chronic inflammatory skin disease characterized by dysregulation of the interleukin 17 (IL-17) signaling axis. Given that psoriasis development depends on keratinocyte stem cells and early progenitors' sensitivity to differentiation, we analyzed IL-17 ligands and the expression and function of in a novel subset of keratinocyte subpopulations: keratinocyte stem cells (KSC) and early and late Transit Amplifying (ETA or LTA, respectively) cells. We found that all subpopulations expressed all IL-17 variants, predominantly in ETA and LTA. Conversely, IL-17 receptor expression resulted in more heterogeneity, with IL-17RA, -C, and -E being the most differentially regulated. Stimulus with IL-17A, IL-17-F, IL-17-A/F, and IL-17C promotes the upregulation of CXCL1, CXCL8, and DEFB4 mRNAs expression in both KSC and ETA. Moreover, IL-17A and IL-17A/F mainly decrease KSC proliferation and promote cell cycle block. Globally, IL-17A and IL-17A/F modulated the expression of proliferation, differentiation, and psoriasis-associated markers. Furthermore, KSC- and ETA-derived 3D reconstructions displayed increased epidermal thickness and upregulated KRT16 expression after treatment with IL-17A or IL-17A/F. Therefore, our data demonstrated that IL-17 family members perform distinctive functions in a specific keratinocyte subpopulation and define IL-17 signaling as a critical modulator of KSC behavior, proving its role in epidermal homeostasis dysregulation of psoriasis.
Collapse
Affiliation(s)
- Elisabetta Palazzo
- DermoLab, Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy; (R.L.); (M.Q.); (C.P.); (A.M.)
| | | | | | | | | |
Collapse
|
3
|
Rastrick J, Edwards H, Ferecskó AS, Le Friec G, Manghera A, Page M, Shaw S. The roles of interleukin (IL)-17A and IL-17F in hidradenitis suppurativa pathogenesis: evidence from human in vitro preclinical experiments and clinical samples. Br J Dermatol 2025; 192:660-671. [PMID: 39531733 DOI: 10.1093/bjd/ljae442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/02/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Hidradenitis suppurativa (HS) is a chronic relapsing inflammatory skin disease associated with significant comorbidities and poor quality of life. Despite uncertainty about pathways driving inflammation in HS lesions, the cytokines interleukin (IL)-17A and IL-17F have been shown to be upregulated in patients with HS. Previous studies have demonstrated that the monoclonal IgG1 antibody bimekizumab selectively inhibits IL-17F in addition to IL-17A. OBJECTIVES To further investigate the roles of IL-17A and IL-17F in HS pathogenesis. METHODS RNA sequencing (RNAseq) was conducted on skin biopsies taken at baseline and after treatment at week 12 of a phase II proof-of-concept study of bimekizumab in patients with moderate-to-severe HS. Differentially expressed genes were identified between baseline lesional and nonlesional samples and between lesional samples before and after bimekizumab treatment, to describe molecular disease mechanisms and treatment effect. Human hair follicular keratinocytes (HHFK) were cultured and treated with a supernatant of stimulated T helper (Th)17 cells in combination with anti-IL-17A, anti-IL-17F, anti-IL-17A and anti-IL-17F, or IgG control antibodies. Total mRNA was analysed by RNAseq. Cellular supernatants from the stimulated HHFKs were used as a source of Th17-induced chemoattractants in neutrophil chemotaxis assays. RESULTS RNAseq revealed that the most prominently upregulated genes in HS lesions included those associated with neutrophil biology. Bimekizumab treatment resulted in reduced expression of these genes. The extent of reduction in gene expression was dependent on achieving HiSCR50 (≥ 50% reduction from baseline in the total abscess and inflammatory nodule count, with no increase from baseline in abscess or draining tunnel count). In vitro dual inhibition of IL-17A and IL-17F had greater attenuation of Th17-induced HS-associated genes and neutrophil migration in HHFKs vs. IL-17A or IL-17F inhibition alone. In situ hybridization found that IL-17A- and IL-17F-producing cells in HS lesions can lack the IL-23 receptor and IL-1β could induce IL-23-independent IL-17F expression in vitro. Furthermore, mucosal-associated invariant cells in HS tunnels expressed IL-17F and IL-1 receptor type 1. IL-1β-, IL-17A- and IL-17F-expressing cells were found to be co-localized in HS lesions. CONCLUSIONS These data support the hypothesis that IL-17A and IL-17F play central roles in HS, a neutrophilic dermatosis. The presence of IL-1β may partly explain the high expression of IL-17F in lesional HS tissue.
Collapse
|
4
|
Untaaveesup S, Amnartpanich T, Leelakanok N, Owattanapanich W, Kantagowit P, Dendumrongsup W, Sornwanee S, Srichana P, Pratchyapruit W, Laisuan W. The prevalence and risk of allergic rhinitis in psoriasis patients: a systematic review and meta-analysis. Sci Rep 2025; 15:9009. [PMID: 40089525 PMCID: PMC11910509 DOI: 10.1038/s41598-025-86779-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 01/14/2025] [Indexed: 03/17/2025] Open
Abstract
Psoriasis, a chronic inflammatory systemic disease, may increase the risk of allergic diseases. This meta-analysis assesses the prevalence and risk of developing allergic rhinitis in psoriasis. We systematically searched MEDLINE, Scopus, and EMBASE for population-based studies documenting AR in psoriasis compared to those without from inception to December 2023. Meta-analysis was conducted using the random-effects model. Eight studies, comprising 5 cross-sectional studies, 1 case-control study, 1 retrospective cohort study, and 1 prospective cohort study, were included. The increased prevalence of AR was 22.29% (95% CI: 0.135 to 0.281; P < 0.001; I2 = 99.05%) in the psoriasis population. The AR risk in psoriasis was insignificant (adjusted OR 1.19%; 95% CI: 0.69 to 2.06; I2 = 99%; P < 0.00001). Subgroup analysis in moderate to severe psoriasis cases revealed a trend of higher risk of AR (OR 1.41%; 95% CI: 0.94 to 2.10; I2 = 92%; P = 0.0003) with very low certainty of evidence. High heterogeneity was observed in most analyses. Our meta-analysis demonstrated a higher prevalence of AR in psoriasis patients. Evaluating and treating allergic diseases can enhance holistic treatment.
Collapse
Affiliation(s)
| | - Thipsukon Amnartpanich
- Department of Ophthalmology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nattawut Leelakanok
- Division of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Burapha University, Chonburi, Thailand
| | - Weerapat Owattanapanich
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Center of Excellence of Siriraj Adult Acute Myeloid/Lymphoblastic Leukemia, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | | | | | | | | | - Wannada Laisuan
- Division of Allergy Immunology and Rheumatology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand, 10400, Thailand.
| |
Collapse
|
5
|
An S, Cui J, Yang W, Zhang M, Yu H, Lu J, Tian Y, Qiao L, Wang X, Bao L, Zhao P. HAS-CIRCpedia-5280 sponges miR-4712-5p inhibited colon cancer autophagyinduced by human beta-defensin-1. J Transl Med 2025; 23:281. [PMID: 40050987 PMCID: PMC11883960 DOI: 10.1186/s12967-024-05860-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 11/06/2024] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND Among all malignancies, colorectal cancer ranks third in incidence rate and second in mortality rate. Human beta-defensin-1 (hBD-1) has broad-spectrum antimicrobial properties, and it plays an important role in the tumor microenvironment. Circular ribonucleic acids (circRNAs) regulate the proliferation and progression of colorectal cancer cells via cancer-related signaling pathways. METHODS Cell proliferation was assessed using the Cell Counting Kit-8 assay to determine the optimal hBD-1 concentration. Intracellular autophagic vesicles were visualized via monodansylcadaverine staining. In addition, the levels of AKT and mammalian target of rapamycin (mTOR)-associated signaling proteins were analyzed via Western blot analysis. CircRNA microarrays and quantitative real-time polymerase chain reaction were used to identify differentially expressed circRNAs in colon cancer cell lines. The functional role of HAS-CIRCpedia-5280 in vitro was demonstrated by overexpressing HAS-CIRCpedia-5280 and inhibiting miR-4712-5p. HAS-CIRCpedia-5280 could be a sponge of miR-4712-5p, mimicking the effect induced by HAS-CIRCpedia-5280 overexpression in colon cancer cells. RESULTS hBD-1 inhibited the proliferation of colon cancer cells and increased the number of intracellular autophagic vesicles. In addition, hBD-1 inhibited the AKT/mTOR signaling pathway, thereby enhancing cellular autophagy. Further, the interaction of HAS-CIRCpedia-5280 and miR-4712-5p was investigated. hBD-1 upregulated the expression level of HAS-CIRCpedia-5280 and downregulated the expression level of miR-4712-5p in colon cancer cells. Subsequently, the overexpression of HAS-CIRCpedia-5280 or the inhibition of miR-4712-5p activated the AKT/mTOR signaling pathway, leading to cellular autophagy inhibition. Conversely, the mimicry of miR-4712-p counteracted the effect of HAS-CIRCpedia 5280 overexpression in colon cancer cells by inhibiting the activation of the AKT/mTOR signaling pathway and, thereby, enhancing cellular autophagy. CONCLUSION hBD-1 can have an inhibitory effect against cell proliferation in colon cancer SW-620/HCT-116 cells via the HAS-CIRCpedia-5280/miR-4712-5p-mediated activation of autophagy.
Collapse
Affiliation(s)
- Shixiang An
- Laboratory of Microbiology and Immunology, School of Basic Medical Science, Inner Mongolia Medical University, Xinhua Street, Hohhot, 010059, PR China
| | - Jiaxian Cui
- Laboratory of Microbiology and Immunology, School of Basic Medical Science, Inner Mongolia Medical University, Xinhua Street, Hohhot, 010059, PR China
| | - Wenhong Yang
- Laboratory of Microbiology and Immunology, School of Basic Medical Science, Inner Mongolia Medical University, Xinhua Street, Hohhot, 010059, PR China
| | - Mingyu Zhang
- Laboratory of Microbiology and Immunology, School of Basic Medical Science, Inner Mongolia Medical University, Xinhua Street, Hohhot, 010059, PR China
| | - Huiling Yu
- Laboratory of Microbiology and Immunology, School of Basic Medical Science, Inner Mongolia Medical University, Xinhua Street, Hohhot, 010059, PR China
| | - Jingkun Lu
- Laboratory of Microbiology and Immunology, School of Basic Medical Science, Inner Mongolia Medical University, Xinhua Street, Hohhot, 010059, PR China
| | - Yunpeng Tian
- Laboratory of Microbiology and Immunology, School of Basic Medical Science, Inner Mongolia Medical University, Xinhua Street, Hohhot, 010059, PR China
| | - Lu Qiao
- Laboratory of Microbiology and Immunology, School of Basic Medical Science, Inner Mongolia Medical University, Xinhua Street, Hohhot, 010059, PR China
| | - Xiumei Wang
- Medical Oncology, Affiliated People's Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010020, China.
| | - Lili Bao
- Laboratory of Microbiology and Immunology, School of Basic Medical Science, Inner Mongolia Medical University, Xinhua Street, Hohhot, 010059, PR China.
| | - Pengwei Zhao
- Laboratory of Microbiology and Immunology, School of Basic Medical Science, Inner Mongolia Medical University, Xinhua Street, Hohhot, 010059, PR China.
| |
Collapse
|
6
|
Goedken ER, Su Z, Lipovsky A, Kannan A, Chu KL, Ciura S, Foley SE, Frank KE, Goess CA, Gopalakrishnan S, Greszler SN, Khan HA, Leys LJ, King JJ, Mathieu SL, Panchal SC, Paulsboe S, Perham M, Ramos AL, Slivka PF, Srikumaran M, Webster MP, Wambeke EL, Zhu H, Scott VE, McGaraughty S, Honore P. Small molecule interleukin (IL) 17A/A antagonists and antibodies blocking both IL17A/A and IL17A/F demonstrate equivalent degrees of efficacy in preclinical models of skin and joint inflammation. J Pharmacol Exp Ther 2025; 392:103525. [PMID: 40127522 DOI: 10.1016/j.jpet.2025.103525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 02/21/2025] [Indexed: 03/26/2025] Open
Abstract
T-helper 17 (Th17) cells produce homodimeric IL17A/A and IL17F/F cytokines as well as the heterodimeric IL17A/F isoform, all having well known roles in defense against extracellular pathogens including fungal infection. Antibodies targeting IL17A (such as secukinumab and ixekizumab) have been approved to treat psoriasis, psoriatic arthritis, ankylosing spondylitis, and axial spondyloarthritis and are under further investigation as therapies in inflammatory disorders such as hidradenitis suppurativa and giant cell arteritis. Because many patients dislike injections with needles, orally bioavailable small molecule IL17 antagonists are desirable as next-generation drugs as long as they can replicate the degree of efficacy observed with anti-IL17A biologics. We recently described novel small molecules binding as 2 copies to the IL17A/A homodimer with only weak effects on the IL17A/F heterodimer. Because approved antibodies binding IL17A neutralize both IL17A/A and IL17A/F, we assessed whether targeting IL17A/A would be sufficient to bring efficacy comparable to IL17A biologics. In comparison to IL17A/F and IL17F/F, we found that the IL17A/A homodimer is the strongest initiator of signaling and that comparable IL17A/A to IL17A/F ratios are expressed in Th17 cells and in human psoriatic skin tissue. Furthermore, our IL17A/A-specific small molecules block the effects of Th17 cell supernatants on keratinocytes to similar maximal responses as anti-IL17A. Our IL17A/A-selective antagonists deliver comparable efficacy to anti-IL17A biologics in several rodent inflammatory models of psoriasiform dermatitis and arthritis. These results support neutralizing IL17A/A with oral small molecule antagonists is an attractive approach to provide differentiated, next-generation therapies for inflammatory disorders. SIGNIFICANCE STATEMENT: This study found that orally active small molecule antagonists of the proinflammatory cytokine IL17A that preferentially bind the IL17A/A form produced equivalent efficacy to monoclonal antibodies that can neutralize both IL17A/A and IL17A/F. This indicates that the IL17A/A homodimer is the dominant isoform driving inflammation in diseases such as psoriasis and that oral inhibitors targeting IL17A/A may be useful next-generation IL17 therapeutics.
Collapse
Affiliation(s)
| | - Zhi Su
- AbbVie Inc, North Chicago, Illinois
| | | | | | | | | | - Sage E Foley
- AbbVie Bioresearch Center, Worcester, Massachusetts
| | | | | | | | | | | | | | - Jacob J King
- AbbVie Bioresearch Center, Worcester, Massachusetts
| | | | | | | | - Matt Perham
- AbbVie Bioresearch Center, Worcester, Massachusetts
| | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Teo LTK, Juantuah-Kusi N, Subramanian G, Sampath P. Psoriasis Treatments: Emerging Roles and Future Prospects of MicroRNAs. Noncoding RNA 2025; 11:16. [PMID: 39997616 PMCID: PMC11858470 DOI: 10.3390/ncrna11010016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/22/2025] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
Psoriasis, a widespread and chronic inflammatory skin disorder, is marked by its persistence and the lack of a definitive cure. The pathogenesis of psoriasis is increasingly understood, with ongoing research highlighting the intricate interplay of genetic, immunological, and environmental factors. Recent advancements have illuminated the pivotal role of microRNAs in orchestrating complex processes in psoriasis and other hyperproliferative skin diseases. This narrative review highlights the emerging significance of miRNAs as key regulators in psoriasis pathogenesis and examines their potential as therapeutic targets. We discuss current treatment approaches and the promising future of miRNAs as next-generation therapeutic agents for this condition.
Collapse
Affiliation(s)
- Li Tian Keane Teo
- Department of Life Sciences, Imperial College London, Sir Ernst Chain Building, South Kensington, London SW7 2AZ, UK
| | - Nerissa Juantuah-Kusi
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology, and Research (A*STAR), 8A Biomedical Grove #06-06 Immunos, Singapore 138648, Singapore
| | - Gowtham Subramanian
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology, and Research (A*STAR), 8A Biomedical Grove #06-06 Immunos, Singapore 138648, Singapore
- Skin Research Institute of Singapore (SRIS), 11 Mandalay Road #17-01 Clinical Sciences Building, Singapore 308232, Singapore
| | - Prabha Sampath
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology, and Research (A*STAR), 8A Biomedical Grove #06-06 Immunos, Singapore 138648, Singapore
- Skin Research Institute of Singapore (SRIS), 11 Mandalay Road #17-01 Clinical Sciences Building, Singapore 308232, Singapore
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School 8 College Road, Singapore 169857, Singapore
| |
Collapse
|
8
|
Zhang J, Shi X, Bai G, Chen J, Fu Y, Chen A. Association between the exposure to brominated flame retardants and psoriasis risk among U.S. adults: A population-based study. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 291:117836. [PMID: 39919589 DOI: 10.1016/j.ecoenv.2025.117836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 01/12/2025] [Accepted: 01/31/2025] [Indexed: 02/09/2025]
Abstract
Psoriasis risk can be affected by exposure to mixtures of environmental chemicals; however, the effect of brominated flame retardants (BFRs) exposure on self-reported psoriasis has not been assessed. Herein, we evaluated the association between the exposure to BFRs compounds of 6 chemicals with risk of psoriasis. Based on data from the National Health and Nutrition Examination Survey (NHANES) 2003-2006 and 2009-2014, this analysis was conducted among 6340 adults. Multivariable logistic regression, restricted cubic splines (RCS), weighted quantile sum (WQS) regression, quantile-based g computation (Qgcomp) regression, and Bayesian kernel machine regression (BKMR) models were performed to estimate the associations. Our results indicated that co-exposure to BFRs was associated with increased risk of psoriasis. Notably, PBB153 emerged as a significant factor contributing to the risk of psoriasis, and showed a non-linear relationship between the concentration of serum PBB153 and the prevalence of psoriasis. Moreover, the association between PBB153 and psoriasis risk remained stable across all subgroup analyses, including those stratified by gender. Our study presents robust evidence connecting BFRs exposure to the prevalence of psoriasis, emphasizing the necessity for continued research and policy interventions to address this environmental health issue. Because BFRs are extensively used and detected in various environmental sources and living organisms, future investigations should further elucidate the mechanisms underlying the observed associations between BFRs exposure and psoriasis risk.
Collapse
Affiliation(s)
- Jingbo Zhang
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Xiangyun Shi
- College of Geography and Resources, Sichuan Normal University, Chengdu 610101, China; Key Laboratory of Land Resources Evaluation and Monitoring in Southwest, Ministry of Education, Sichuan Normal University, Chengdu 610101, China
| | - Genlong Bai
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jin Chen
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yidian Fu
- Graduate School of Hebei Medical University, Shijiazhuang, Hebei 050017, China; Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei 050051, China.
| | - Aijun Chen
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| |
Collapse
|
9
|
Merzel Šabović EK, Kraner Šumenjak T, Božič Mijovski M, Janić M. Residual non-specific and disease-specific inflammatory markers in successfully treated young psoriasis patients: a cross-sectional study. Immunol Res 2025; 73:28. [PMID: 39775226 PMCID: PMC11711139 DOI: 10.1007/s12026-024-09584-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025]
Abstract
Psoriasis is a chronic, immune-mediated disease. The systemic inflammation triggered by psoriasis contributes significantly to increased cardiovascular risk. While various treatments completely clear the skin, the associated effects on systemic inflammation are not yet clear. We investigated residual systemic inflammation in successfully treated patients. Circulating disease-specific and non-specific inflammatory markers were measured and compared in 80 psoriasis patients (aged 30-45 years) successfully treated with topical therapy, methotrexate, adalimumab, secukinumab or guselkumab, and in 20 healthy controls. Non-specific inflammatory markers (high-sensitivity C-reactive protein (hs-CRP), complete blood count (CBC) parameters, neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), mean platelet volume-to-platelet ratio (MPR), and red blood cell distribution width-to-platelet ratio (RPR)) and disease-specific inflammatory markers (interferon-γ (IFN-γ), tumor necrosis factor (TNF), interleukin (IL)-1β, IL-12p70, IL-17, and IL-23) were measured and compared between groups. Disease-specific cytokines (IFN-γ, TNF, IL-1β, IL-12p70, and IL-17, but not IL-23), were significantly elevated in patients compared to controls, while non-specific inflammatory markers showed no differences compared to controls. The residual disease-specific cytokines were similarly elevated in all five treated groups. In addition, they correlated significantly with body mass index (BMI) and waist circumference. Our results suggest that psoriasis patients have elevated residual disease-specific cytokines despite successful treatment, while the non-specific inflammatory markers are similar to those in control subjects. Residual disease-specific inflammatory markers correlated with BMI and waist circumference. A possible beneficial effect of body weight control in psoriasis patients merits further investigation. The study was registered at http://clinicaltrials.gov (identifier: NCT05957120) on July 24, 2023.
Collapse
Affiliation(s)
- Eva Klara Merzel Šabović
- Department of Dermatovenerology, University Medical Centre Ljubljana, Gradiškova ulica 10, Ljubljana, Slovenia.
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana, Slovenia.
| | - Tadeja Kraner Šumenjak
- Faculty of Agriculture and Life Sciences, University of Maribor, Pivola 10, Hoče, Slovenia
| | - Mojca Božič Mijovski
- Department of Vascular Diseases, Laboratory for Haemostasis and Atherothrombosis, University Medical Centre Ljubljana, Zaloška 7, Ljubljana, Slovenia
| | - Miodrag Janić
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana, Slovenia
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, Zaloška 7, Ljubljana, Slovenia
| |
Collapse
|
10
|
Abhale K, Veeranjaneyulu A, Desai S. A Snapshot of Biomarkers in Psoriasis. Curr Drug Discov Technol 2025; 22:e180324228068. [PMID: 38500289 DOI: 10.2174/0115701638278470240312075112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 02/16/2024] [Accepted: 02/26/2024] [Indexed: 03/20/2024]
Abstract
A persistent long-standing, inflammatory skin condition that is brought on by a variety of factors is psoriasis. It is distinguished by itchy, scaly, reddish plaques, particularly on areas of the body that are frequently chafed, including the extensor sites of the limbs. Recent developments in molecular-targeted therapy that use biologics or small-molecule inhibitors can effectively cure even the worst psoriatic indications. The outstanding clinical outcomes of treatment help to clarify the disease's detrimental consequences on quality of life. Biomarkers that identify deep remission are essential for developing uniform treatment plans. Blood protein markers such as AMPs that are consistently quantifiable can be very helpful in routine clinical practice. The metabolic pathways involve biomarkers that can not only help diagnose psoriasis in a clinical setting but also indicate its severity based on the levels present in the body. Machine learning and AI have made a diagnosis of the expression of genes as biomarkers more accessible. In this article, biomarkers, as well as their key role in psoriasis, are discussed.
Collapse
Affiliation(s)
- Krushna Abhale
- Department of Pharmacology, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, India
| | | | - Shivani Desai
- Clinical Research and Pharmacovigilance, Serum Institute of India Pvt. Ltd., Hadapsar, Pune, India
| |
Collapse
|
11
|
Maldonado-García JL, Fragozo A, Pavón L. Cytokine release syndrome induced by anti-programmed death-1 treatment in a psoriasis patient: A dark side of immune checkpoint inhibitors. World J Clin Cases 2024; 12:6782-6790. [PMID: 39687650 PMCID: PMC11525914 DOI: 10.12998/wjcc.v12.i35.6782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/26/2024] [Accepted: 09/09/2024] [Indexed: 10/24/2024] Open
Abstract
In recent years, cancer immunotherapy has introduced novel treatments, such as monoclonal antibodies, which have facilitated targeted therapies against tumor cells. Programmed death-1 (PD-1) is an immune checkpoint expressed in T cells that regulates the immune system's activity to prevent over-activation and tissue damage caused by inflammation. However, PD-1 is also expressed in tumor cells and functions as an immune evasion mechanism, making it a therapeutic target to enhance the immune response and eliminate tumor cells. Consequently, immune checkpoint inhibitors (ICIs) have emerged as an option for certain tumor types. Nevertheless, blocking immune checkpoints can lead to immune-related adverse events (irAEs), such as psoriasis and cytokine release syndrome (CRS), as exemplified in the clinical case presented by Zhou et al involving a patient with advanced gastric cancer who received sintilimab, a monoclonal antibody targeting PD-1. Subsequently, the patient experienced exacerbation of psoriasis and CRS. The objective of this editorial article is to elucidate potential immunologic mechanisms that may contribute to the development of CRS and psoriasis in patients receiving ICIs. It is crucial to acknowledge that while ICIs offer superior safety and efficacy compared to conventional therapies, they can also manifest irAEs affecting the skin, gastrointestinal tract, or respiratory system. In severe cases, these irAEs can lead to life-threatening complications such as circulatory shock or multiorgan failure. Consequently, it is recommended that patients receiving ICIs undergo regular monitoring to identify and manage these adverse events effectively.
Collapse
Affiliation(s)
- José Luis Maldonado-García
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacán 04510, Ciudad de México, Mexico
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Mexico City 1134, Ciudad de México, Mexico
| | - Ana Fragozo
- Unidad de Desarrollo e Investigación en Bioterapéuticos, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Ciudad de México, Mexico
| | - Lenin Pavón
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 11340, Mexico
| |
Collapse
|
12
|
Desai R, Irwin C, McCune M, Davis M. Racial disparities in length of hospitalization and systemic medication utilization in patients with psoriasis. Arch Dermatol Res 2024; 317:106. [PMID: 39666078 DOI: 10.1007/s00403-024-03623-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 08/27/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024]
Abstract
The growing evidence of chronic inflammation and sequelae caused by psoriasis indicates the need for systemic treatment, including biologics and conventional treatments like methotrexate. However, unconscious racial bias may lead to discrepancies in systemic medication prescription. Racial identity is also suspected to affect comorbidity and hospitalization rates in patients with psoriasis. This study examines the impact of race on hospitalization duration and systemic medication prescription patterns in patients with psoriasis in the United States in 2020. The 2020 Healthcare Cost and Utilization (HCUP) National Inpatient Sample (NIS) database was used, with inpatient admissions over the age of 17 with an ICD-10-coded psoriasis diagnosis included. Patient demographics and hospital characteristics were compared between "non-white" and "white" patients, with white patients as the reference group. The primary outcome, systemic medication prescription, was evaluated using logistic regression models, producing an odds ratio and corresponding 95% confidence interval (CI). The secondary outcome, length of stay, was evaluated using negative binomial regression models to approximate the incident rate ratio and 95% CI. 1,802,720 patients with psoriasis were included in this study. Hispanic, Asian or Pacific Islander, and Native American patients with psoriasis had significantly lower odds of systemic medication prescription than white patients with psoriasis. All non-white racial identities were found to have significantly longer estimated lengths of stay compared to white patients with psoriasis. Disparities in the medication of choice for psoriasis treatment can lead to increased inpatient burden with longer lengths of stay for non-white patients.
Collapse
Affiliation(s)
- Rini Desai
- University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA.
| | - Chase Irwin
- University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Mariana McCune
- University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Mitchell Davis
- University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| |
Collapse
|
13
|
Hagino T, Onda M, Saeki H, Fujimoto E, Kanda N. Effectiveness of bimekizumab for genital, nail, and scalp lesions with psoriasis: A 24-week real-world study. J Dermatol 2024; 51:1658-1664. [PMID: 39133570 DOI: 10.1111/1346-8138.17427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/02/2024] [Accepted: 07/28/2024] [Indexed: 12/08/2024]
Abstract
Psoriasis is a complex, chronic inflammatory skin disease that significantly impacts patients' quality of life (QOL), especially in cases of genital, nail, and scalp psoriasis. Bimekizumab is an inhibitor of interleukin (IL)-17A and IL-17F and used for the treatment of psoriasis. The aim of this retrospective study was to evaluate the effectiveness of bimekizumab through in treating genital, nail, and scalp lesions with psoriasis over 24 weeks. The study was conducted from May 2022 and February 2024 on 52 psoriasis patients treated with bimekizumab. The therapeutic effects of bimekizumab were evaluated by the achievement of Physician's Global Assessment (PGA) rates of 0/1 on the genitals (genital-PGA), fingernails (PGA-F), scalp-specific PGA (ss-PGA), static PGA (sPGA), and the Dermatology Life Quality Index (DLQI) at weeks 4, 16, and 24. Bimekizumab treatment significantly improved genital, nail, and scalp lesions with psoriasis. At week 24, the achievement rate of genital-PGA 0/1, PGA-F 0/1, ss-PGA 0/1 was 96.2%, 66.7%, or 93.9%, and that of sPGA 0/1 or DLQI 0/1 was 93.9% or 83.3%, respectively. Bimekizumab was effective for genital, nail, and scalp lesions with psoriasis, difficult-to-treat lesions, and simultaneously improved QOL in a real-world clinical practice.
Collapse
Affiliation(s)
- Teppei Hagino
- Department of Dermatology, Nippon Medical School Chiba Hokusoh Hospital, Inzai, Japan
| | - Marina Onda
- Department of Dermatology, Nippon Medical School Chiba Hokusoh Hospital, Inzai, Japan
| | - Hidehisa Saeki
- Department of Dermatology, Nippon Medical School, Tokyo, Japan
| | | | - Naoko Kanda
- Department of Dermatology, Nippon Medical School Chiba Hokusoh Hospital, Inzai, Japan
| |
Collapse
|
14
|
Luengas‐Martinez A, Ismail D, Paus R, Young HS. Vascular endothelial growth factor A inhibition remodels the transcriptional signature of lipid metabolism in psoriasis non-lesional skin in 12 h ex vivo culture. SKIN HEALTH AND DISEASE 2024; 4:e471. [PMID: 39624732 PMCID: PMC11608907 DOI: 10.1002/ski2.471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/09/2024] [Accepted: 10/11/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Vascular endothelial growth factor A (VEGF-A)-mediated angiogenesis is involved in the pathogenesis of psoriasis. VEGF-A inhibitors are widely used to treat oncological and ophthalmological diseases but have not been used in psoriasis management. The molecular mechanisms underlying the effects of VEGF-A inhibition in psoriatic skin remain unknown. OBJECTIVES To identify the genes and canonical pathways affected by VEGF-A inhibition in non-lesional and plaque skin ex vivo. METHODS Total RNA sequencing was performed on skin biopsies from patients with psoriasis (n = 6; plaque and non-lesional skin) and healthy controls (n = 6) incubated with anti-VEGF-A monoclonal antibody (bevacizumab, Avastin®) or human IgG1 isotype control for 12 h in serum-free organ culture. Differentially expressed genes between paired control and treated samples with adjusted p-values <0.1 were considered significant. Gene ontology and ingenuity pathway analysis was used to identify enriched biological processes, canonical pathways and upstream regulators. RESULTS VEGF-A inhibition upregulated the expression of genes involved in lipid metabolism. Pathway enrichment analysis identified the activation of pathways involved in fatty acids and lipid biosynthesis and degradation in non-lesional skin and ferroptosis in plaque skin. VEGF-A inhibition downregulated endothelial cell apoptosis in non-lesional psoriasis skin and members of the interferon family were identified as potential regulators of the effects of VEGF-A inhibition in non-lesional skin. CONCLUSION Early response to VEGF-A inhibition is associated with changes in lipid metabolism in non-lesional psoriasis skin and cellular stress in psoriasis plaque. More investigation is needed to validate these findings.
Collapse
Affiliation(s)
- Andrea Luengas‐Martinez
- Centre for Dermatology Research and Manchester Academic Health Science CentreThe University of ManchesterManchesterUK
| | - Dina Ismail
- Centre for Dermatology Research and Manchester Academic Health Science CentreThe University of ManchesterManchesterUK
| | - Ralf Paus
- Centre for Dermatology Research and Manchester Academic Health Science CentreThe University of ManchesterManchesterUK
- Dr. Philip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Monasterium LaboratoryMuensterGermany
| | - Helen S. Young
- Centre for Dermatology Research and Manchester Academic Health Science CentreThe University of ManchesterManchesterUK
| |
Collapse
|
15
|
Morgner B, Werz O, Wiegand C, Tittelbach J. Bilayered skin equivalent mimicking psoriasis as predictive tool for preclinical treatment studies. Commun Biol 2024; 7:1529. [PMID: 39558145 PMCID: PMC11574237 DOI: 10.1038/s42003-024-07226-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 11/07/2024] [Indexed: 11/20/2024] Open
Abstract
Psoriasis is a prevalent, inflammatory skin disease without cure. Further research is required to unravel dysregulated processes and develop new therapeutic interventions. The lack of suitable in vivo and in vitro preclinical models is an impediment in the psoriasis research. Recently, the development of 3D skin models has progressed including replicas with disease-like features. To investigate the use of in vitro models as preclinical test tools, the study focused on treatment responses of 3D skin replicas. Cytokine-priming of skin organoids induced psoriatic features like inflammation, antimicrobial peptides (AMP), hyperproliferation and impaired differentiation. Topical application of dexamethasone (DEX) or celastrol (CEL), a natural anti-inflammatory compound reduced the secretion of pro-inflammatory cytokines. DEX and CEL decreased the gene expression of inflammatory mediators. DEX barely affected the psoriatic AMP transcription but CEL downregulated psoriasis-driven AMP genes. Subcutaneous application of adalimumab (ADM) or bimekizumab (BMM) showed anti-psoriatic effects via protein induction of the differentiation marker keratin-10. Dual blockage of TNF-α and IL-17A repressed the inflammatory psoriasis phenotype. BMM inhibited the psoriatic expression of AMP genes and induced KRT10 and cell-cell contact genes. The present in vitro model provides a 3D environment with in vivo-like cutaneous responses and represents a promising tool for preclinical investigations.
Collapse
Affiliation(s)
- Bianka Morgner
- University Hospital Jena, Department of Dermatology, Friedrich Schiller University Jena, Jena, Germany.
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Cornelia Wiegand
- University Hospital Jena, Department of Dermatology, Friedrich Schiller University Jena, Jena, Germany
| | - Jörg Tittelbach
- University Hospital Jena, Department of Dermatology, Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
16
|
Sortebech D, Schoenfeldt T, Duvetorp A, Agerholm-Nielsen R, Eidsmo L. Skin-resident T Cells Contribute to the Dynamic Disease Manifestations of Psoriasis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1267-1277. [PMID: 39432869 DOI: 10.4049/jimmunol.2400020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 08/23/2024] [Indexed: 10/23/2024]
Abstract
The human skin forms a dynamic barrier to physical injuries and microbial invasion. Constant interactions between stroma and tissue-confined immune cells maintain skin homeostasis. However, the cellular interactions that maintain skin health also contribute to focal immunopathology. Psoriasis is a common disease that manifests with focal pathology induced by environmental triggers in genetically susceptible individuals. Within psoriasis plaques, cross-talk between skin-resident T cells and stroma cells leads to chronic inflammation. Inflammatory cytokines such as TNF-α, IL-17, IL-22, and IL-23 amplify the local chronic inflammation and sustain the well-demarcated thick and scaly plaques that characterize the disease. In resolved lesions, T cells remain poised for IL-17 and IL-22 production, and postinflammatory epigenetic modifications lower the threshold for initiation of local relapse. This review focuses on how tissue-resident memory T cells contribute to the onset, maintenance, resolution, and relapse of psoriasis.
Collapse
Affiliation(s)
- Daniel Sortebech
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Trine Schoenfeldt
- Leo Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
| | - Albert Duvetorp
- Leo Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
- Department of Dermatology, Skåne University Hospital, Malmö, Sweden
| | - Rasmus Agerholm-Nielsen
- Leo Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
| | - Liv Eidsmo
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Leo Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
17
|
Lin L, Luo Q, Gao X, Li Q, Li W, Zhou X, Liu W, Zhong X, Yang Y, Zhang X. Correlation and Clinical Significance of HBD-2 and CXCL-1/2 Levels at Skin Lesions with Psoriasis Vulgaris Severity. Immunol Invest 2024; 53:1234-1249. [PMID: 39301953 DOI: 10.1080/08820139.2024.2395852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
OBJECTIVE This study was performed to explore the clinical significance of the expression of human beta-defensin 2 (HBD-2) and chemokine ligand 1/2 (CXCL-1/2) in psoriasis vulgaris. METHODS This study retrospectively included the study group (n = 160) and control group (n = 100) for analysis. The levels of inflammatory indicators, blood biochemical indicators, and immune indicators using ELISA. The psoriasis area and severity index (PASI) was used to evaluate disease severity. Levels of HBD-2, CXCL-1, CXCL-2 and CCL20 were determined by RT-PCR. The correlations of HBD-2, CXCL-1 and CXCL-2 levels with CCL20 and PASI scores were analyzed. The diagnostic value of HBD-2, CXCL-1 and CXCL-2 in psoriasis vulgaris was analyzed by ROC curve. RESULTS HBD-2, CXCL-1 and CXCL-2 were highly expressed in the lesions of psoriasis vulgaris patients, and were positively correlated with CCL20 and PASI score. HBD-2, CXCL-1 and CXCL-2 alone or in combination had high diagnostic value for psoriasis vulgaris and severe psoriasis, and the combined diagnostic value of the three was higher than that of a single indicator. CONCLUSION HBD-2, CXCL-1, and CXCL-2 levels are closely related to the severity of psoriasis vulgaris and can effectively diagnose the occurrence and progression of psoriasis vulgaris.
Collapse
Affiliation(s)
- Ling Lin
- Department of Dermatology, Guangzhou Dermatology Hospital, Guangzhou, China
| | - Quan Luo
- Department of Dermatology, Guangzhou Dermatology Hospital, Guangzhou, China
| | - Xinjing Gao
- Department of Dermatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Qian Li
- Department of Dermatology, Guangzhou Dermatology Hospital, Guangzhou, China
| | - Wei Li
- Department of Dermatology, Guangzhou Dermatology Hospital, Guangzhou, China
| | - Xin Zhou
- Department of Dermatology, Guangzhou Dermatology Hospital, Guangzhou, China
| | - Weiyu Liu
- Department of Dermatology, Guangzhou Dermatology Hospital, Guangzhou, China
| | - Xuelian Zhong
- Department of Dermatology, Guangzhou Dermatology Hospital, Guangzhou, China
| | - Yunqing Yang
- Department of Dermatology, Guangzhou Dermatology Hospital, Guangzhou, China
| | - Xibao Zhang
- Department of Dermatology, Guangzhou Dermatology Hospital, Guangzhou, China
| |
Collapse
|
18
|
Adams R, Bunick CG, Lawson ADG, Gomez B, Shaw S. Crystal Structure of Bimekizumab Fab Fragment in Complex with IL-17F Provides Molecular Basis for Dual IL-17A and IL-17F Inhibition. J Invest Dermatol 2024; 144:2581-2583.e2. [PMID: 38631666 DOI: 10.1016/j.jid.2024.03.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/19/2024] [Accepted: 03/22/2024] [Indexed: 04/19/2024]
Affiliation(s)
| | - Christopher G Bunick
- Department of Dermatology, Yale School of Medicine, Yale University, New Haven, Connecticut, USA; Program in Translational Biomedicine, Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| | | | | | | |
Collapse
|
19
|
Campione E, Artosi F, Shumak RG, Giunta A, Argenziano G, Assorgi C, Balato A, Bernardini N, Brunasso AMG, Burlando M, Caldarola G, Campanati A, Carugno A, Castelli F, Conti A, Costanzo A, Cuccia A, Dapavo P, Dattola A, De Simone C, Di Lernia V, Dini V, Donini M, Errichetti E, Esposito M, Fargnoli MC, Foti A, Fiorella C, Gargiulo L, Gisondi P, Guarneri C, Legori A, Lembo S, Loconsole F, Malagoli P, Marzano AV, Mercuri SR, Megna M, Micali G, Mortato E, Musumeci ML, Narcisi A, Offidani AM, Orsini D, Paolino G, Pellacani G, Peris K, Potenza C, Prignano F, Quaglino P, Ribero S, Richetta AG, Romanelli M, Rossi A, Strippoli D, Trovato E, Venturini M, Bianchi L. Fast Clinical Response of Bimekizumab in Nail Psoriasis: A Retrospective Multicenter 36-Week Real-Life Study. Pharmaceuticals (Basel) 2024; 17:1378. [PMID: 39459016 PMCID: PMC11510175 DOI: 10.3390/ph17101378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/19/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
(1) Background/Objectives: Nail psoriasis (NP) is a chronic and difficult-to-treat disease, which causes significant social stigma and impairs the patients' quality of life. Moreover, nail psoriasis is a true therapeutic challenge for clinicians. The presence of nail psoriasis can be part of a severe form of psoriasis and can have predictive value for the development of psoriatic arthritis. Our real-world-evidence multicenter study aims to evaluate the efficacy of bimekizumab in nail psoriasis. (2) Methods: A retrospective analysis of a multicenter observational study included 834 patients affected by moderate-to-severe psoriasis, in 33 Dermatologic Units in Italy, treated with bimekizumab from December 2022 to September 2023. Clinimetric assessments were based on Psoriasis Area and Severity Index (PASI), Dermatology Life Quality Index (DLQI), and Physician's Global Assessment of Fingernail Psoriasis (PGA-F) for the severity of nail psoriasis at 0, 12, 24, and 36 weeks. (3) Results: Psoriatic nail involvement was present in 27.95% of patients. The percentage of patients who achieved a complete clearance of NP in terms of PGA-F 0 was 31.7%, 57%, and 88.5% at week 4, 16, and 36, respectively. PASI 100 was achieved by 32.03% of patients at week 4, by 61.8% at week 16, and by 78.92% of patients at week 36. The mean baseline PASI was 16.24. The mean DLQI values for the entire group of patients at baseline, at week 4, at week 16, and at week 36 were 14.62, 3.02, 0.83, and 0.5, respectively. (4) Conclusions: Therapies that promote the healing of both the skin and nails in a short time can also ensure a lower risk of subsequently developing arthritis which is disabling over time. Bimekizumab proved to be particularly effective to treat NP, with a fast response in terms of complete clearance, with over 88.5% of patients free from NP after 36 weeks. The findings of our real-world study showed that patients with moderate-to-severe PsO and concomitant NP had significantly faster and more substantial improvements in NP up to 36 weeks with respect to previous research findings. Considering the rapid healing of the nail, the dual inhibition of IL17 A and F might have a great value in re-establishing the dysregulation of keratin 17 at the nail level.
Collapse
Affiliation(s)
- Elena Campione
- Dermatology Unit, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (F.A.); (R.G.S.); (A.G.); (L.B.)
| | - Fabio Artosi
- Dermatology Unit, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (F.A.); (R.G.S.); (A.G.); (L.B.)
| | - Ruslana Gaeta Shumak
- Dermatology Unit, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (F.A.); (R.G.S.); (A.G.); (L.B.)
| | - Alessandro Giunta
- Dermatology Unit, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (F.A.); (R.G.S.); (A.G.); (L.B.)
| | - Giuseppe Argenziano
- Dermatology Unit, University of Campania L. Vanvitelli, 80131 Naples, Italy; (G.A.); (A.B.)
| | - Chiara Assorgi
- Daniele Innocenzi, Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University Dermatology ASL, 04100 Latina, Italy; (C.A.); (N.B.); (C.P.)
| | - Anna Balato
- Dermatology Unit, University of Campania L. Vanvitelli, 80131 Naples, Italy; (G.A.); (A.B.)
| | - Nicoletta Bernardini
- Daniele Innocenzi, Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University Dermatology ASL, 04100 Latina, Italy; (C.A.); (N.B.); (C.P.)
| | | | - Martina Burlando
- Dermatological Clinic, Department of Clinical and Molecular Sciences, Polytechnic Marche University, 60100 Ancona, Italy; (M.B.); (A.C.); (A.M.O.)
| | - Giacomo Caldarola
- Dermatology, Department of Medicine and Translational Surgery, Università Cattolica del Sacro Cuore, 00185 Rome, Italy; (G.C.); (C.D.S.); (K.P.)
- Dermatology, Department of Medical and Surgery Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Anna Campanati
- Dermatological Clinic, Department of Clinical and Molecular Sciences, Polytechnic Marche University, 60100 Ancona, Italy; (M.B.); (A.C.); (A.M.O.)
| | - Andrea Carugno
- Dermatology Unit, Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy;
| | - Franco Castelli
- Section of Dermatology, Koelliker Hospital, 47923 Turin, Italy; (F.C.); (A.C.)
| | - Andrea Conti
- Section of Dermatology, Koelliker Hospital, 47923 Turin, Italy; (F.C.); (A.C.)
| | - Antonio Costanzo
- Dermatology Unit, IRCCS Humanitas Research Hospital, 10134 Rozzano, Italy; (A.C.); (L.G.)
| | - Aldo Cuccia
- Unit of Dermatology, San Donato Hospital, 52100 Arezzo, Italy;
| | - Paolo Dapavo
- Second Dermatologic Clinic, Department of Biomedical Science and Human Oncology, University of Turin, 10124 Turin, Italy;
| | - Annunziata Dattola
- Dermatology Unit, Department of Clinical Internal, Anesthesiological and Cardiovascular Science, University of La Sapienza, 00161 Rome, Italy; (A.D.); (G.P.); (A.G.R.); (A.R.)
| | - Clara De Simone
- Dermatology, Department of Medicine and Translational Surgery, Università Cattolica del Sacro Cuore, 00185 Rome, Italy; (G.C.); (C.D.S.); (K.P.)
- Dermatology, Department of Medical and Surgery Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Vito Di Lernia
- Dermatology Unit, Arcispedale Santa Maria Nuova, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy;
| | - Valentina Dini
- Dermatology Unit, Department of Clinical and Experimental Medicine Ospedale Santa Chiara, 56126 Pisa, Italy; (V.D.); (M.R.)
| | - Massimo Donini
- Dermatology Unit, Department of Medicine, Hospital S.S. Giovanni e Paolo, AULSS−3-Serenissima, 30122 Venezia, Italy;
| | - Enzo Errichetti
- Institute of Dermatology, Department of Medicine, University of Udine, 33100 Udine, Italy;
| | - Maria Esposito
- Section of Dermatology, Department of Biotechnological and Applied Clinical Science, University of L’Aquila, 67100 L’Aquila, Italy; (M.E.); (M.C.F.)
| | - Maria Concetta Fargnoli
- Section of Dermatology, Department of Biotechnological and Applied Clinical Science, University of L’Aquila, 67100 L’Aquila, Italy; (M.E.); (M.C.F.)
| | - Antonio Foti
- Unit of Dermatology, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (A.F.); (S.R.M.); (G.P.)
| | - Carmen Fiorella
- Section of Dermatology, Oncology and Ematology Department Asl Bat, P.O. M.R. Dimiccoli, 70051 Barletta, Italy;
| | - Luigi Gargiulo
- Dermatology Unit, IRCCS Humanitas Research Hospital, 10134 Rozzano, Italy; (A.C.); (L.G.)
| | - Paolo Gisondi
- Department of Medicine, Section of Dermatology and Venereology, University of Verona, 37129 Verona, Italy;
| | - Claudio Guarneri
- Department of Biomedical and Dental Sciences and Morpho Functional Imaging, Section of Dermatology, University of Messina, 98121 Verona, Italy;
| | - Agostina Legori
- UO Dermatologia IRCCS Ospedale Galeazzi & Università degli Studi di Milano, 20157 Milan, Italy;
| | - Serena Lembo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana” University of Salerno, 84084 Salerno, Italy;
| | - Francesco Loconsole
- Department of Dermatology, University of Bari, 70121 Bari, Italy; (F.L.); (E.M.)
| | - Piergiorigio Malagoli
- Department of Dermatology, Dermatology Unit Azienda Ospedaliera San Donato Milanese, 20097 Milan, Italy;
| | - Angelo Valerio Marzano
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy
| | - Santo Raffaele Mercuri
- Unit of Dermatology, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (A.F.); (S.R.M.); (G.P.)
- Unit of Dermatologic Clinic, Università Vita-Salute, San Raffaele, 20132 Milan, Italy
| | - Matteo Megna
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, 80138 Naple, Italy;
| | - Giuseppe Micali
- UOC Dermatologia, University of Catania, PO “G. Rodolico”, AOU Policlinico “G. Rodolico-San Marco”, 95123 Catania, Italy; (G.M.); (M.L.M.)
| | - Edoardo Mortato
- Department of Dermatology, University of Bari, 70121 Bari, Italy; (F.L.); (E.M.)
| | - Maria Letizia Musumeci
- UOC Dermatologia, University of Catania, PO “G. Rodolico”, AOU Policlinico “G. Rodolico-San Marco”, 95123 Catania, Italy; (G.M.); (M.L.M.)
| | - Alessandra Narcisi
- Dermatology Unit, IRCCS Humanitas Research Hospital, 10134 Rozzano, Italy; (A.C.); (L.G.)
| | - Anna Maria Offidani
- Dermatological Clinic, Department of Clinical and Molecular Sciences, Polytechnic Marche University, 60100 Ancona, Italy; (M.B.); (A.C.); (A.M.O.)
| | - Diego Orsini
- Clinical Dermatology Unit, San Gallicano Dermatological Institute IRCCS, 00167 Rome, Italy;
| | - Giovanni Paolino
- Unit of Dermatology, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (A.F.); (S.R.M.); (G.P.)
| | - Giovanni Pellacani
- Dermatology Unit, Department of Clinical Internal, Anesthesiological and Cardiovascular Science, University of La Sapienza, 00161 Rome, Italy; (A.D.); (G.P.); (A.G.R.); (A.R.)
| | - Ketty Peris
- Dermatology, Department of Medicine and Translational Surgery, Università Cattolica del Sacro Cuore, 00185 Rome, Italy; (G.C.); (C.D.S.); (K.P.)
- Dermatology, Department of Medical and Surgery Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Concetta Potenza
- Daniele Innocenzi, Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University Dermatology ASL, 04100 Latina, Italy; (C.A.); (N.B.); (C.P.)
| | - Francesca Prignano
- Department of Dermatological Sciences, Dermatology Section, University of Florence, 50121 Florence, Italy;
| | - Pietro Quaglino
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Turin, Torino, Italy; (P.Q.); (S.R.)
| | - Simone Ribero
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Turin, Torino, Italy; (P.Q.); (S.R.)
| | - Antonio Giovanni Richetta
- Dermatology Unit, Department of Clinical Internal, Anesthesiological and Cardiovascular Science, University of La Sapienza, 00161 Rome, Italy; (A.D.); (G.P.); (A.G.R.); (A.R.)
| | - Marco Romanelli
- Dermatology Unit, Department of Clinical and Experimental Medicine Ospedale Santa Chiara, 56126 Pisa, Italy; (V.D.); (M.R.)
| | - Antonio Rossi
- Dermatology Unit, Department of Clinical Internal, Anesthesiological and Cardiovascular Science, University of La Sapienza, 00161 Rome, Italy; (A.D.); (G.P.); (A.G.R.); (A.R.)
| | - Davide Strippoli
- Dermatology Unit, Manzoni Hospital, ASST-Lecco, 23900 Lecco, Italy;
| | - Emanuele Trovato
- Dermatology Unit, Department of Medical, Surgical and Neurological Sciences, University of Siena, 53100 Siena, Italy;
| | - Marina Venturini
- Department of Clinical and Experimental Sciences, Section of Dermatology, University of Brescia, 25123 Brescia, Italy;
| | - Luca Bianchi
- Dermatology Unit, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (F.A.); (R.G.S.); (A.G.); (L.B.)
| |
Collapse
|
20
|
Radaschin DS, Tatu A, Iancu AV, Beiu C, Popa LG. The Contribution of the Skin Microbiome to Psoriasis Pathogenesis and Its Implications for Therapeutic Strategies. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1619. [PMID: 39459406 PMCID: PMC11509136 DOI: 10.3390/medicina60101619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024]
Abstract
Psoriasis is a common chronic inflammatory skin disease, associated with significant morbidity and a considerable negative impact on the patients' quality of life. The complex pathogenesis of psoriasis is still incompletely understood. Genetic predisposition, environmental factors like smoking, alcohol consumption, psychological stress, consumption of certain drugs, and mechanical trauma, as well as specific immune dysfunctions, contribute to the onset of the disease. Mounting evidence indicate that skin dysbiosis plays a significant role in the development and exacerbation of psoriasis through loss of immune tolerance to commensal skin flora, an altered balance between Tregs and effector cells, and an excessive Th1 and Th17 polarization. While the implications of skin dysbiosis in psoriasis pathogenesis are only starting to be revealed, the progress in the characterization of the skin microbiome changes in psoriasis patients has opened a whole new avenue of research focusing on the modulation of the skin microbiome as an adjuvant treatment for psoriasis and as part of a long-term plan to prevent disease flares. The skin microbiome may also represent a valuable predictive marker of treatment response and may aid in the selection of the optimal personalized treatment. We present the current knowledge on the skin microbiome changes in psoriasis and the results of the studies that investigated the efficacy of the different skin microbiome modulation strategies in the management of psoriasis, and discuss the complex interaction between the host and skin commensal flora.
Collapse
Affiliation(s)
- Diana Sabina Radaschin
- Department of Clinical Medical, Faculty of Medicine and Pharmacy, “Saint Parascheva” Infectious Disease Clinical Hospital, Multidisciplinary Integrated Centre of Dermatological Interface Research Centre (MICDIR), “Dunarea de Jos” University of Galati, 800008 Galati, Romania
| | - Alin Tatu
- Department of Clinical Medical, Faculty of Medicine and Pharmacy, “Saint Parascheva” Infectious Disease Clinical Hospital, Multidisciplinary Integrated Centre of Dermatological Interface Research Centre (MICDIR), “Dunarea de Jos” University of Galati, 800008 Galati, Romania
| | - Alina Viorica Iancu
- Department of Morphological and Functional Sciences, “Dunarea de Jos” University of Galati, 800008 Galati, Romania
| | - Cristina Beiu
- Department of Oncologic Dermatology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Liliana Gabriela Popa
- Department of Oncologic Dermatology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
21
|
Catlett IM, Gao L, Hu Y, Banerjee S, Krueger JG. Pharmacodynamic Response to Deucravacitinib, an Oral, Selective, Allosteric TYK2 Inhibitor, in a Global, Phase 2, Randomized, Double-Blind, Placebo-Controlled Psoriasis Trial. Dermatol Ther (Heidelb) 2024; 14:2827-2839. [PMID: 39283417 PMCID: PMC11480296 DOI: 10.1007/s13555-024-01262-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/20/2024] [Indexed: 10/17/2024] Open
Abstract
BACKGROUND Psoriasis, a chronic, immune-mediated, inflammatory disease, affects 2‒3% of the population. Tyrosine kinase 2 (TYK2) mediates cytokine signaling involved in adaptive [interleukin (IL)-12, IL-23] and innate (type-I interferons) immune responses; IL-23-driven T-helper (Th)17 pathways play a key role in chronic inflammation in psoriasis. In a phase 2 trial, deucravacitinib, an oral, selective, allosteric TYK2 inhibitor, reduced IL-23/Th17 and type-I interferon pathway expression in the skin of patients with moderate to severe plaque psoriasis, reductions that were accompanied by clinical improvement of psoriatic lesions. OBJECTIVES The aim of this study was to identify biomarkers of psoriatic disease in serum from patients enrolled in the phase 2 trial and to assess the effects of deucravacitinib on those biomarkers. METHODS Serum biomarkers from Olink proteomics and other quantitative assays were evaluated for a pharmacodynamic response to deucravacitinib treatment and correlation with psoriasis disease activity measures. RESULTS Serum biomarkers associated with the IL-23/Th17 pathway [IL-17A, IL-17C, IL-19, IL-20, beta-defensin, and peptidase inhibitor 3 (PI3)] were upregulated in patients with psoriasis versus healthy controls. Deucravacitinib treatment reduced IL-17A (adjusted mean change from baseline at Day 85; 12 mg once daily versus placebo; -0.240 versus -0.067), IL-17C (-14.850 versus -1.664), IL-19 (-96.445 versus -8.119), IL-20 (-0.265 versus -0.064), beta-defensin (-65,025.443 versus -7553.961), and PI3 (-14.005 versus -1.360) expression. Reductions in serum biomarker expression occurred in a dose- and time-dependent manner, with significant reductions from baseline seen with deucravacitinib doses ≥ 3 mg twice daily (P ≤ 0.05). Biomarker expression correlated with disease activity measures such as Psoriasis Area and Severity Index (PASI) at baseline. Biomarker expression also correlated with PASI scores at Week 12. CONCLUSION IL-23/Th17 pathway expression in the serum of patients with psoriasis is an indicator of disease activity and response to deucravacitinib treatment. TRIAL REGISTRATION NUMBER NCT02931838.
Collapse
Affiliation(s)
- Ian M Catlett
- Bristol Myers Squibb, 3401 Princeton Pike, Lawrenceville, NJ, 08648, USA
| | - Lu Gao
- Bristol Myers Squibb, 3401 Princeton Pike, Lawrenceville, NJ, 08648, USA.
| | - Yanhua Hu
- Bristol Myers Squibb, 3401 Princeton Pike, Lawrenceville, NJ, 08648, USA
| | - Subhashis Banerjee
- Bristol Myers Squibb, 3401 Princeton Pike, Lawrenceville, NJ, 08648, USA
| | | |
Collapse
|
22
|
FitzGerald O, Gladman DD, Mease PJ, Ritchlin C, Smolen JS, Gao L, Hu Y, Nowak M, Banerjee S, Catlett I. Phase 2 Trial of Deucravacitinib in Psoriatic Arthritis: Biomarkers Associated With Disease Activity, Pharmacodynamics, and Clinical Responses. Arthritis Rheumatol 2024; 76:1397-1407. [PMID: 38770592 DOI: 10.1002/art.42921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 05/22/2024]
Abstract
OBJECTIVE Our objective was to evaluate the association of serum biomarkers with baseline psoriatic arthritis (PsA) disease activity, pharmacodynamic effects of deucravacitinib on biomarker levels, and the relationship between biomarkers and clinical responses to deucravacitinib. METHODS The phase 2 trial (ClinicalTrials.gov identifier: NCT03881059) randomly assigned 203 patients with PsA 1:1:1 to placebo, deucravacitinib at 6 mg once daily (QD), or deucravacitinib at 12 mg QD. Serum biomarkers associated with the interleukin 23 (IL-23) pathway (IL-17A, β-defensin [BD-2], and IL-19), type I interferon pathway, inflammation, and collagen matrix turnover were measured by immunoassay. Clinical responses (≥75% improvement from baseline in the Psoriasis Area and Severity Index [PASI75] and ≥20% improvement from baseline in American College of Rheumatology criteria [ACR20] responses) were measured at week 16. Hematologic variables were also assessed. RESULTS IL-17A, BD-2, and IL-19 had a modest association with PASI scores (r = 0.4, r = 0.56, and r = 0.5, respectively) at baseline. In deucravacitinib groups, IL-17A, BD-2, IL-19, C-X-C motif ligand 9 (CXCL9), CXCL10, C-reactive protein, matrix metalloproteinase 3, and collagen type 4 degradation marker levels were significantly reduced at week 16 versus baseline (P < 0.01); higher levels of IL-23 pathway-associated biomarkers predicted higher PASI75 and ACR20 response rates in deucravacitinib-treated patients. Significantly higher PASI75 response rates were seen in patients with high baseline IL-17A (odds ratio 15.76) and BD-2 levels (odds ratio 15.41) versus low baseline IL-17A and BD-2 levels. Changes in hematologic variables that are characteristic of JAK inhibition were not observed with deucravacitinib. CONCLUSION Deucravacitinib significantly impacted biomarkers associated with Tyk2 signaling pathways of key inflammatory cytokines, including IL-23 and type I interferon, and those related to collagen matrix turnover. These biomarkers may predict treatment responses to deucravacitinib.
Collapse
Affiliation(s)
| | | | | | | | - Josef S Smolen
- Medical University of Vienna and Hietzing Hospital, Vienna, Austria
| | - Lu Gao
- Bristol Myers Squibb, Princeton, New Jersey
| | - Yanhua Hu
- Bristol Myers Squibb, Princeton, New Jersey
| | | | | | | |
Collapse
|
23
|
Eyerich K, Asadullah K, Pinter A, Weisenseel P, Reich K, Paul C, Sabat R, Wolk K, Eyerich S, Lauffer F, Angsana J, Taut FJH, Kohler K, Chen Y, Sendecki J, Leung MWL, Wegner S, Personke Y, Gomez M, Krüger N, Tabori S, Schäkel K. Noninferiority of 16-Week vs 8-Week Guselkumab Dosing in Super Responders for Maintaining Control of Psoriasis: The GUIDE Randomized Clinical Trial. JAMA Dermatol 2024; 160:953-963. [PMID: 39083288 PMCID: PMC11292573 DOI: 10.1001/jamadermatol.2024.2463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/04/2024] [Indexed: 08/03/2024]
Abstract
Importance Psoriasis is a chronic inflammatory skin disease with unmet needs for tailored treatment and therapy de-escalation strategies. Objective To evaluate early intervention with and prolonging the dosing interval for guselkumab, a p19 subunit-targeted interleukin (IL)-23 inhibitor, in patients with moderate to severe psoriasis. Design, Setting, and Participants The GUIDE clinical trial is an ongoing phase 3b, randomized, double-blinded trial conducted across 80 centers in Germany and France comprising 3 parts evaluating the impact of early disease intervention, prolonged dosing interval, and maintenance of response following treatment withdrawal among adults with moderate to severe plaque psoriasis. In study part 2, reported herein, first and last patient visits were September 2019 and March 2022, respectively. Interventions In GUIDE part 1 (week [W]0-W28), patients received guselkumab, 100 mg, at W0, W4, W12, and W20. Those achieving a Psoriasis Area and Severity Index (PASI) of 0 at both W20 and W28 were termed super responders (SRes). In part 2 (W28-W68), SRes were randomized to guselkumab, 100 mg, every 8 weeks or every 16 weeks; non-SRes continued open-label guselkumab every 8 weeks. Main Outcomes and Measures Primary objective was to demonstrate noninferiority (with a 10% margin) of guselkumab every 16 weeks vs every 8 weeks dosing among SRes for maintenance of disease control (PASI <3 at W68). Biomarker substudies assessed immunologic effects in skin and blood. Results Overall, 822 patients received guselkumab in part 2 (297 [36.1%] SRes [every 8 weeks/every 16 weeks; n = 148/n = 149] and 525 [63.9%] non-SRes). Among SRes, mean (SD) age was 39.4 (14.1) years, 95 (32.0%) were female, and 202 (68.0%) were male. The primary end point of noninferiority for guselkumab every 16 weeks vs every 8 weeks in SRes was met (P = .001), with 91.9% (137/149; 90% CI, 87.3%-95.3%) of SRes receiving every 16 weeks and 92.6% (137/148; 90% CI, 88.0%-95.8%) of SRes receiving dosing every 8 weeks having PASI lower than 3 at W68. Clinical effects corresponded with immunologic changes; skin CD8-positive tissue-resident memory T (TRM)-cell count decreased quickly from baseline, remaining low in both dosing groups. Similarly, serum IL-17A, IL-17F, IL-22, and β defensin (BD)-2 levels decreased significantly from baseline, remaining low in both dosing groups to W68. Guselkumab was well-tolerated; no new safety signals were identified. Conclusions and Relevance Psoriasis treatment guidelines lack or provide inconsistent advice on patient stratification and treatment de-escalation. We present the first randomized trial providing evidence that, in patients with early complete skin clearance at 2 consecutive visits (W20 and W28), extending the guselkumab dosing interval may control disease activity. Trial Registration ClinicalTrials.gov Identifier: NCT03818035.
Collapse
Affiliation(s)
- Kilian Eyerich
- Department of Dermatology and Venereology, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - Khusru Asadullah
- Department of Dermatology, Venereology and Allergology, Charité – Universitätsmedizin, Berlin, Germany
- Dermatology Potsdam MVZ, Potsdam, Germany
| | - Andreas Pinter
- University Hospital Frankfurt am Main, Frankfurt, Germany
| | | | - Kristian Reich
- Translational Research in Inflammatory Skin Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Robert Sabat
- Psoriasis Research and Treatment Center, Department of Dermatology, Venereology and Allergology, and Institute of Medical Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kerstin Wolk
- Psoriasis Research and Treatment Center, Department of Dermatology, Venereology and Allergology, and Institute of Medical Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Stefanie Eyerich
- Center for Allergy and Environment, Technical University and Helmholtz Center Munich, Munich, Germany
| | - Felix Lauffer
- Center for Allergy and Environment, Technical University and Helmholtz Center Munich, Munich, Germany
| | | | | | | | | | | | | | | | | | - Mario Gomez
- Janssen Global Services LLC, Horsham, Pennsylvania
| | | | | | - Knut Schäkel
- Department of Dermatology, and Interdisciplinary Center for Inflammatory Diseases, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
24
|
Kaur P, Prabhahar A, Pal D, Nada R, Kohli HS, Kumar V, Ramachandran R. IL-23/IL-17 in a Paradoxical Association with Primary Membranous Nephropathy. Inflammation 2024; 47:1536-1544. [PMID: 38393549 DOI: 10.1007/s10753-024-01992-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/20/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024]
Abstract
Primary membranous nephropathy (PMN), an autoimmune disease, is the most common cause of nephrotic syndrome in middle-aged non-diabetic adults. PMN pathophysiology includes Th1/Th2 paradigm. The IL-23/IL-17 pathway is implicated in autoimmune kidney disorders, but no study has examined its relationship with PMN. In several unrelated studies, PMN patients reported to have paradoxical IL-17 levels. This manuscript describes the best possible association of IL-23/IL-17 axis with PMN. Biopsy-proven PMN patients and age, gender-matched healthy controls were enrolled. Serum-PLA2R (Euroimmune, Germany), IL-23 and IL-17 (R&D; USA), was measured using ELISA along with biochemical parameters. Appropriate statistical tools were used for analysis. One hundred eighty-nine PMN patients (mean age 41.70 ± 12.53 years) and 100 controls (mean age 43.92 ± 10.93 years) were identified. One hundred forty were PLA2R-related. PMN patients had median proteinuria, serum albumin, and creatinine of 6.12 (3.875, 9.23) g/day, 2.32 (1.96, 2.9) g/dl, and 0.89 (0.7, 1.1) mg/dl, respectively. IL-17, but not IL-23, was significantly increased in PMN patients compared to controls (IL-17, median: 12.07 pg/ml (9.75, 24.56) vs median: 9.75 pg/ml (8.23, 17.03) p = 0.0002); (IL23, median: 6.04 pg/ml (4.22, 10.82) vs median: 5.46 pg/ml (3.34, 9.96) p = 0.142). IL-17 and IL-23 correlated significantly (p 0.05) in PMN patients, and similar trend was seen when grouped into PLA2R-related and -unrelated groups. The levels of IL-23 (p = 0.057) and IL-17 (p = 0.004) were high in MN patients that did not respond to the treatment. The current finding may indicate or suggest the involvement of IL-23/IL-17 PMN pathogenesis. A comprehensive investigation is needed to evaluate IL-23/IL-17 axis with renal infiltrating immune cells, and external stimuli.
Collapse
Affiliation(s)
- Prabhjot Kaur
- Department of Nephrology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Arun Prabhahar
- Department of Nephrology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Deeksha Pal
- Department of Nephrology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Ritambhra Nada
- Department of Histopathology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Harbir Singh Kohli
- Department of Nephrology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Vinod Kumar
- Department of Dermatology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India.
| | - Raja Ramachandran
- Department of Nephrology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India.
| |
Collapse
|
25
|
Emami Z, Shobeiri SS, Khorrami R, Haghnavaz N, Rezaee MA, Moghadam M, Pordel S, Sankian M. Evaluation of Kynu, Defb2, Camp, and Penk Expression Levels as Psoriasis Marker in the Imiquimod-Induced Psoriasis Model. Mediators Inflamm 2024; 2024:5821996. [PMID: 39045230 PMCID: PMC11265934 DOI: 10.1155/2024/5821996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 06/26/2024] [Indexed: 07/25/2024] Open
Abstract
Background Psoriasis is a noncontagious auto-inflammatory chronic skin disease. So far, some of the inflammatory genes were upregulated in mouse model of psoriasis. This study examined changes in skin mRNA expression of L-kynureninase (Kynu), cathelicidin antimicrobial peptide (Camp), beta-defensin 2 (Defb2), and proenkephalin (Penk) in a mouse model of imiquimod-induced psoriasis. Materials and Methods Tree groups of C57BL/6 female mice were allocated. The imiquimod (IMQ) cream was administered to the mice dorsal skin of the two groups to induce psoriatic inflammation. In the treatment group, IMQ was administered 10 min after hydrogel-containing M7 anti-IL-17A aptamer treatment. Vaseline (Vas) was administered to the negative control group. The psoriatic skin lesions were evaluated based on the psoriasis area severity index (PASI) score, histopathology, and mRNA expression levels of Kynu, Camp, Defb2, and Penk using real-time PCR. In order to assess the systemic response, the spleen and lymph node indexes were also evaluated. Results The PASI and epidermal thickness scores were 6.01 and 1.96, respectively, in the IMQ group, and they significantly decreased after aptamer administration to 1.15 and 0.90, respectively (P < 0.05). Spleen and lymph node indexes showed an increase in the IMQ group, followed by a slight decrease after aptamer treatment (P > 0.05). Additionally, the mRNA expression levels of Kynu, Defb2, Camp, and Penk genes in the IMQ-treated region showed a significant 2.70, 4.56, 3.29, and 2.61-fold increase relative to the Vas mice, respectively (P < 0.05). The aptamer-treated region exhibited a significant decrease in these gene expression levels (P < 0.05). A positive correlation was found between Kynu, Penk, and Camp expression levels and erythema, as well as Camp expression with PASI, scaling, and thickness (P < 0.05). Conclusion According to our results, it seems that Kynu, Camp, and Penk can be considered appropriate markers for the evaluation of psoriasis in IMQ-induced psoriasis. Also, the anti-IL-17 aptamer downregulated these important genes in this mouse model.
Collapse
Affiliation(s)
- Zahra Emami
- Immunology Research CenterFaculty of MedicineMashhad University of Medical Sciences, Mashhad, Iran
| | - Saeideh Sadat Shobeiri
- Immunology Research CenterFaculty of MedicineMashhad University of Medical Sciences, Mashhad, Iran
- Cellular and Molecular Research CenterSabzevar University of Medical Sciences, Sabzevar, Iran
| | - Razia Khorrami
- Immunology Research CenterFaculty of MedicineMashhad University of Medical Sciences, Mashhad, Iran
| | - Navideh Haghnavaz
- Immunology Research CenterFaculty of MedicineMashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ali Rezaee
- Immunology Research CenterFaculty of MedicineMashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Laboratory SciencesFaculty of ParamedicalKurdistan University of Medical Sciences, Sanandaj, Iran
| | - Malihe Moghadam
- Immunology Research CenterFaculty of MedicineMashhad University of Medical Sciences, Mashhad, Iran
| | - Safoora Pordel
- Immunology Research CenterFaculty of MedicineMashhad University of Medical Sciences, Mashhad, Iran
| | - Mojtaba Sankian
- Immunology Research CenterFaculty of MedicineMashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
26
|
Alsabbagh MM. Cytokines in psoriasis: From pathogenesis to targeted therapy. Hum Immunol 2024; 85:110814. [PMID: 38768527 DOI: 10.1016/j.humimm.2024.110814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/22/2024]
Abstract
Psoriasis is a multifactorial disease that affects 0.84% of the global population and it can be associated with disabling comorbidities. As patients present with thick scaly lesions, psoriasis was long believed to be a disorder of keratinocytes. Psoriasis is now understood to be the outcome of the interaction between immunological and environmental factors in individuals with genetic predisposition. While it was initially thought to be solely mediated by cytokines of type-1 immunity, namely interferon-γ, interleukin-2, and interleukin-12 because it responds very well to cyclosporine, a reversible IL-2 inhibitor; the discovery of Th-17 cells advanced the understanding of the disease and helped the development of biological therapy. This article aims to provide a comprehensive review of the role of cytokines in psoriasis, highlighting areas of controversy and identifying the connection between cytokine imbalance and disease manifestations. It also presents the approved targeted treatments for psoriasis and those currently under investigation.
Collapse
Affiliation(s)
- Manahel Mahmood Alsabbagh
- Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders and Department of Molecular Medicine, Arabian Gulf University, Manama, Bahrain.
| |
Collapse
|
27
|
Sintès M, Kovjenic P, Haine (Hablal) L, Serror K, Beladjine M, Parietti (Montcuquet) V, Delagrange M, Ducos B, Bouaziz JD, Boccara D, Mimoun M, Bensussan A, Bagot M, Huang N, Michel L. Coencapsulation of Immunosuppressive Drug with Anti-Inflammatory Molecule in Pickering Emulsions as an Innovative Therapeutic Approach for Inflammatory Dermatoses. JID INNOVATIONS 2024; 4:100273. [PMID: 39045393 PMCID: PMC11264173 DOI: 10.1016/j.xjidi.2024.100273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 01/11/2024] [Accepted: 02/13/2024] [Indexed: 07/25/2024] Open
Abstract
Psoriasis is an inflammatory skin disease characterized by epidermal and immune dysfunctions. Although efficient, current topical treatments display adverse effects, including skin atrophy and burning sensation, leading to poor patient adherence. To overcome these downsides, pickering emulsions were formulated in which the calcitriol-containing dispersed phase was stabilized with either cyclosporin A- or tacrolimus-loaded poly(lactic-co-glycolic) acid nanoparticles. This study aimed to investigate their biological effects on lymphocytes and epidermal cells and their effectiveness in an imiquimod-induced psoriasis-like mouse model. Results showed that both emulsions significantly inhibited nuclear factor of activated T cell translocation in T lymphocytes as well as their IL-2 production, cell activation, and proliferation. In keratinocytes, inhibition of nuclear factor of activated T cell translocation decreased the production of IL-8 and TNF-α. Topical application of emulsions over skin biopsies ex vivo showed accumulation of rhodamin B-coupled poly(lactic-co-glycolic) acid nanoparticles throughout the epidermis by immunofluorescence and significantly decreased the antigen-presenting capacity of Langerhans cells in relation to a reduced expression of activation markers CD40, CD86, and HLA-DR. Using an imiquimod-induced psoriasis model in vivo, pickering emulsions significantly alleviated psoriasiform lesions potentially attributed to the decreased cutaneous expression of T-cell markers, proinflammatory cytokines, chemokines, and specific epidermal cell genes. Altogether, pickering emulsion might be a very efficient formulation for treating inflammatory dermatoses.
Collapse
Affiliation(s)
- Maxime Sintès
- Inserm UMR_U976, University Paris Cité, Skin Research Center, Hôpital Saint-Louis, Paris, France
| | - Petra Kovjenic
- University Paris Saclay, CNRS, Institut Galien Paris-Saclay, Orsay, France
| | - Liasmine Haine (Hablal)
- Inserm UMR_U976, University Paris Cité, Skin Research Center, Hôpital Saint-Louis, Paris, France
| | - Kevin Serror
- Department of Reconstructive and Plastic Surgery, Hôpital Saint-Louis, Paris, France
| | - Mohamed Beladjine
- University Paris Saclay, CNRS, Institut Galien Paris-Saclay, Orsay, France
| | | | - Marine Delagrange
- High Throughput qPCR Core Facility, École Normale Supérieure, Université Paris Sciences & Lettres, Paris, France
| | - Bertrand Ducos
- High Throughput qPCR Core Facility, École Normale Supérieure, Université Paris Sciences & Lettres, Paris, France
| | - Jean-David Bouaziz
- Inserm UMR_U976, University Paris Cité, Skin Research Center, Hôpital Saint-Louis, Paris, France
- Department of Dermatology, Hôpital Saint-Louis, Paris, France
| | - David Boccara
- Department of Reconstructive and Plastic Surgery, Hôpital Saint-Louis, Paris, France
| | - Maurice Mimoun
- Department of Reconstructive and Plastic Surgery, Hôpital Saint-Louis, Paris, France
| | - Armand Bensussan
- Inserm UMR_U976, University Paris Cité, Skin Research Center, Hôpital Saint-Louis, Paris, France
| | - Martine Bagot
- Inserm UMR_U976, University Paris Cité, Skin Research Center, Hôpital Saint-Louis, Paris, France
- Department of Dermatology, Hôpital Saint-Louis, Paris, France
| | - Nicolas Huang
- University Paris Saclay, CNRS, Institut Galien Paris-Saclay, Orsay, France
| | - Laurence Michel
- Inserm UMR_U976, University Paris Cité, Skin Research Center, Hôpital Saint-Louis, Paris, France
- Department of Dermatology, Hôpital Saint-Louis, Paris, France
| |
Collapse
|
28
|
Guo Y, Luo L, Zhu J, Li C. Advance in Multi-omics Research Strategies on Cholesterol Metabolism in Psoriasis. Inflammation 2024; 47:839-852. [PMID: 38244176 DOI: 10.1007/s10753-023-01961-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 11/29/2023] [Accepted: 12/25/2023] [Indexed: 01/22/2024]
Abstract
The skin is a complex and dynamic organ where homeostasis is maintained through the intricate interplay between the immune system and metabolism, particularly cholesterol metabolism. Various factors such as cytokines, inflammatory mediators, cholesterol metabolites, and metabolic enzymes play crucial roles in facilitating these interactions. Dysregulation of this delicate balance contributes to the pathogenic pathways of inflammatory skin conditions, notably psoriasis. In this article, we provide an overview of omics biomarkers associated with psoriasis in relation to cholesterol metabolism. We explore multi-omics approaches that reveal the communication between immunometabolism and psoriatic inflammation. Additionally, we summarize the use of multi-omics strategies to uncover the complexities of multifactorial and heterogeneous inflammatory diseases. Finally, we highlight potential future perspectives related to targeted drug therapies and research areas that can advance precise medicine. This review aims to serve as a valuable resource for those investigating the role of cholesterol metabolism in psoriasis.
Collapse
Affiliation(s)
- Youming Guo
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, Jiangsu, China
| | - Lingling Luo
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu, China
| | - Jing Zhu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu, China
| | - Chengrang Li
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu, China.
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, Jiangsu, China.
| |
Collapse
|
29
|
Siebert S, Coates LC, Schett G, Raychaudhuri SP, Chen W, Gao S, Seridi L, Chakravarty SD, Shawi M, Lavie F, Sharaf M, Zimmermann M, Kollmeier AP, Xu XL, Rahman P, Mease PJ, Deodhar A. Modulation of Interleukin-23 Signaling With Guselkumab in Biologic-Naive Patients Versus Tumor Necrosis Factor Inhibitor-Inadequate Responders With Active Psoriatic Arthritis. Arthritis Rheumatol 2024; 76:894-904. [PMID: 38253404 DOI: 10.1002/art.42803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/08/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024]
Abstract
OBJECTIVE We assessed and compared immunologic differences and associations with clinical response to guselkumab, a fully human interleukin (IL)-23p19 subunit inhibitor, in participants with active psoriatic arthritis (PsA) who were biologic-naive or had inadequate response to tumor necrosis factor inhibitors (TNFi-IR). METHODS Serum biomarker levels at baseline and after treatment with guselkumab 100 mg every 8 weeks were compared between biologic-naive (n = 251) and TNFi-IR (n = 93) subgroups identified in the pooled DISCOVER-1/DISCOVER-2/COSMOS data set. Baseline biomarker levels determined by achievement of week 24 clinical responses (≥75%/90% improvement in Psoriasis Area and Severity Index [PASI 75/90], Investigator's Global Assessment [IGA] of psoriasis score 0/1 and ≥2-point improvement], ≥20% improvement in American College of Rheumatology criteria [ACR20]) were compared between prior treatment subgroups. RESULTS Baseline IL-22, TNFα, and beta defensin-2 (BD-2) levels were significantly lower in biologic-naive than in TNFi-IR participants. With guselkumab, week 24 IL-17A, IL-17F, IL-22, serum amyloid A, C-reactive protein, IL-6, and BD-2 levels were significantly reduced from baseline in biologic-naive and TNFi-IR participants (≥1.4-fold difference, nominal P < 0.05). Clinical responders to guselkumab exhibited significantly higher baseline levels of several biomarkers than nonresponders (IL-17A, IL-17F, BD-2 in biologic-naive PASI 90 responders; IL-17A, BD-2 in TNFi-IR IGA 0/1 responders; IL-22, BD-2 in TNFi-IR PASI 90 responders [nominal P < 0.05]) and trended higher in TNFi-IR ACR20 responders. CONCLUSION Guselkumab modulates IL-23 signaling and provides consistent pharmacodynamic effects in both biologic-naive and TNFi-IR PsA patients. Significantly elevated baseline IL-22, TNFα, and BD-2 levels and associations between baseline IL-22, IL-17A, and BD-2 levels and skin responses to guselkumab suggest greater dysregulation of IL-23/Th17 signaling in patients with TNFi-IR.
Collapse
Affiliation(s)
| | | | - Georg Schett
- Friedrich-Alexander Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Siba P Raychaudhuri
- University of California Davis and Veterans Affairs Northern California Health Care System, Mather, California
| | - Warner Chen
- Janssen Research & Development, LLC, Spring House, Pennsylvania
| | - Sheng Gao
- Janssen Research & Development, LLC, Spring House, Pennsylvania
| | - Loqmane Seridi
- Janssen Research & Development, LLC, Spring House, Pennsylvania
| | - Soumya D Chakravarty
- Janssen Scientific Affairs, LLC, Horsham, Pennsylvania, and Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - May Shawi
- Janssen Research & Development, LLC, Titusville, New Jersey
| | - Frederic Lavie
- Global Medical Affairs, Janssen Pharmaceutical Companies of Johnson & Johnson, Issy les Moulineaux, France
| | | | | | | | - Xie L Xu
- Janssen Research & Development, LLC, San Diego, California
| | - Proton Rahman
- Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Philip J Mease
- Swedish Medical Center/Providence St. Joseph Health and University of Washington, Seattle, Washington
| | - Atul Deodhar
- Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
30
|
Maier CS, Eckert D, Laroux FS, Hew KM, Suleiman AA, Liu W, Mohamed MEF. Cedirogant Population Pharmacokinetics and Pharmacodynamic Analyses of Interleukin-17A Inhibition in Two Phase 1 Studies in Healthy Participants and Participants with Moderate to Severe Psoriasis. Clin Pharmacol Drug Dev 2024; 13:474-484. [PMID: 38231873 DOI: 10.1002/cpdd.1377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/19/2023] [Indexed: 01/19/2024]
Abstract
Cedirogant (ABBV-157) is an orally bioavailable inverse agonist of retinoic acid-related orphan receptor gamma thymus. Data from 2 Phase 1 studies were used to characterize cedirogant pharmacokinetics and evaluate target engagement. Cedirogant plasma concentrations and ex vivo interleukin 17A (IL-17A) concentrations from healthy participants and participants with moderate to severe psoriasis (PsO) were analyzed in a population pharmacokinetic and pharmacodynamic modeling framework to characterize cedirogant pharmacokinetics following single and multiple doses and assess ex vivo IL-17A inhibition in relation to cedirogant exposure. Cedirogant population pharmacokinetics were best described by a 2-compartment pharmacokinetic model with delayed absorption and an enzyme turnover compartment to describe cytochrome P450 3A autoinduction. The pharmacokinetics of cedirogant were comparable between healthy participants and participants with PsO. Cedirogant steady-state average and maximum plasma concentrations were predicted to be 7.56 and 11.8 mg/L, respectively, for participants with PsO for the 375 mg once-daily regimen on Day 14. The apparent clearance and apparent volume of distribution for cedirogant were estimated to be 24.5 L/day and 28.2 L, respectively. A direct maximum inhibition model adequately characterized the exposure-response relationship of cedirogant and ex vivo IL-17A inhibition, indicating no temporal delay between exposure and response with a saturable inhibition of IL-17A. Model-estimated half-maximal inhibitory concentration and maximum inhibition values for cedirogant inhibition of ex vivo IL-17A were 0.56 mg/L and 0.76, respectively. The established relationship between cedirogant exposure and biomarker effect supported dose selection for the Phase 2 dose-ranging study in patients with PsO.
Collapse
Affiliation(s)
| | | | | | - Kinjal M Hew
- Precision Medicine Immunology, AbbVie Inc., South San Francisco, CA, USA
| | | | - Wei Liu
- Clinical Pharmacology, AbbVie Inc., North Chicago, IL, USA
| | | |
Collapse
|
31
|
Hsu CY, Yousif AM, Abullah KA, Abbas HH, Ahmad H, Eldesoky GE, Adil M, Hussein Z. Antimicrobial Peptides (AMPs): New Perspectives on Their Function in Dermatological Diseases. Int J Pept Res Ther 2024; 30:33. [DOI: 10.1007/s10989-024-10609-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2024] [Indexed: 01/05/2025]
|
32
|
Kiełbowski K, Stańska W, Bakinowska E, Rusiński M, Pawlik A. The Role of Alarmins in the Pathogenesis of Rheumatoid Arthritis, Osteoarthritis, and Psoriasis. Curr Issues Mol Biol 2024; 46:3640-3675. [PMID: 38666958 PMCID: PMC11049642 DOI: 10.3390/cimb46040228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/13/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Alarmins are immune-activating factors released after cellular injury or death. By secreting alarmins, cells can interact with immune cells and induce a variety of inflammatory responses. The broad family of alarmins involves several members, such as high-mobility group box 1, S100 proteins, interleukin-33, and heat shock proteins, among others. Studies have found that the concentrations and expression profiles of alarmins are altered in immune-mediated diseases. Furthermore, they are involved in the pathogenesis of inflammatory conditions. The aim of this narrative review is to present the current evidence on the role of alarmins in rheumatoid arthritis, osteoarthritis, and psoriasis. We discuss their potential involvement in mechanisms underlying the progression of these diseases and whether they could become therapeutic targets. Moreover, we summarize the impact of pharmacological agents used in the treatment of these diseases on the expression of alarmins.
Collapse
Affiliation(s)
- Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.); (M.R.)
| | - Wiktoria Stańska
- Department of Medical Biology, Medical University of Warsaw, 00-575 Warsaw, Poland;
| | - Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.); (M.R.)
| | - Marcin Rusiński
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.); (M.R.)
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.); (M.R.)
| |
Collapse
|
33
|
Bernardini N, Ambrosio L, Tolino E, Proietti I, Skroza N, Potenza C. Successful Treatment with Bimekizumab of a Psoriatic Patient Undergoing Hemodialysis: A Case Report and Review of the Literature. J Clin Med 2024; 13:2250. [PMID: 38673523 PMCID: PMC11050806 DOI: 10.3390/jcm13082250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Background/Objectives: Treating psoriasis patients requires the consideration of potential underlying complications like latent viral infections and chronic kidney disease, which may influence therapy selection. Case presentation: A patient with end-stage kidney disease (ESKD) undergoing hemodialysis (HD) was successfully treated with bimekizumab, an IgG1 humanized monoclonal antibody inhibiting interleukin (IL)-17A and IL-17F. This case appears to be the first documented instance of effective anti-IL-17A/IL-17F antibody treatment in a psoriasis patient undergoing HD, with a sustained positive response for eight months. Discussion: Studies indicate the comparable pharmacokinetics, efficacy, and safety of certain psoriasis drugs in patients with chronic kidney disease (CKD) and those with normal renal function. The positive clinical outcome observed following treatment with bimekizumab aligns with the existing literature on this topic. However, further studies are needed to objectively evaluate the pharmacokinetics, efficacy, and safety of this drug in this specific setting. Conclusions: This documented case represents the first known use of bimekizumab to treat psoriasis in patients undergoing dialysis, suggesting its potential effectiveness and safety in this population.
Collapse
Affiliation(s)
- Nicoletta Bernardini
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, “Sapienza” University of Rome, Polo Pontino, 04100 Latina, Italy; (N.B.); (E.T.); (I.P.); (N.S.); (C.P.)
| | - Luca Ambrosio
- Dermatology Unit, Department of Clinical Internal Anesthesiologic Cardiovascular Sciences, “Sapienza” University of Rome, 00185 Rome, Italy
| | - Ersilia Tolino
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, “Sapienza” University of Rome, Polo Pontino, 04100 Latina, Italy; (N.B.); (E.T.); (I.P.); (N.S.); (C.P.)
| | - Ilaria Proietti
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, “Sapienza” University of Rome, Polo Pontino, 04100 Latina, Italy; (N.B.); (E.T.); (I.P.); (N.S.); (C.P.)
| | - Nevena Skroza
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, “Sapienza” University of Rome, Polo Pontino, 04100 Latina, Italy; (N.B.); (E.T.); (I.P.); (N.S.); (C.P.)
| | - Concetta Potenza
- Department of Medical-Surgical Sciences and Biotechnologies, Dermatology Unit “Daniele Innocenzi”, “Sapienza” University of Rome, Polo Pontino, 04100 Latina, Italy; (N.B.); (E.T.); (I.P.); (N.S.); (C.P.)
| |
Collapse
|
34
|
Pantoja CJ, Li H, Rodante J, Keel A, Sorokin AV, Svedbom A, Teague HL, Stahle M, Mehta NN, Playford MP. Serum Beta-Defensin-2 is a biomarker for psoriasis but not subclinical atherosclerosis: Role of IL17a, PI-3 kinase and Rac1. JEADV CLINICAL PRACTICE 2024; 3:150-159. [PMID: 38646149 PMCID: PMC11031204 DOI: 10.1002/jvc2.278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 09/20/2023] [Indexed: 04/23/2024]
Abstract
Background Beta-defensins (BDs) are antimicrobial peptides secreted upon epithelial injury. Both chemotactic and antimicrobial properties of BDs function as initial steps in host defense and prime the adaptive immune system in the body. Psoriasis, a chronic immune-mediated inflammatory disease, has both visible cutaneous manifestations as well as known associations with higher incidence of cardiometabolic complications and vascular inflammation. Objectives We aimed to investigate the circulating expression of beta-defensin-2 (BD2) in psoriasis at baseline compared to control subjects, along with changes in BD2 levels following biologic treatment at one-year. The contribution of BD2 to subclinical atherosclerosis is also assessed. In addition, we have sought to unravel signaling mechanisms linking inflammation with BD2 expression. Methods Multimodality imaging as well inflammatory biomarker assays were performed in biologic naïve psoriasis (n=71) and non-psoriasis (n=53) subjects. A subset of psoriasis patients were followed for one-year after biological intervention (anti-Tumor Necrosis Factor-α (TNFα), n=30; anti-Interleukin17A (IL17A), n=21). Measurements of circulating BD2 were completed by Enzyme-Linked Immunosorbent Assay (ELISA). Using HaCaT transformed keratinocytes, expression of BD2 upon cytokine treatment was assessed by quantitative polymerase chain reaction (qPCR) and ELISA. Results Herein, we confirm that human circulating BD2 levels associate with psoriasis, which attenuate upon biologic interventions (anti-TNFα, anti-IL-17A). A link between circulating BD2 and sub-clinical atherosclerosis markers was not observed. Furthermore, we demonstrate that IL-17A-driven BD2 expression occurs in a Phosphatidylinositol 3-kinase (PI3-kinase) and Rac1 GTPase-dependent manner. Conclusions Our findings expand on the potential role of BD2 as a tractable biomarker in psoriasis patients and describes the role of an IL-17A-PI3-kinase/Rac signaling axis in regulating BD2 levels in keratinocytes.
Collapse
Affiliation(s)
- CJ. Pantoja
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung and Blood Institute, National Institute of Health, Bethesda, Maryland
| | - H. Li
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung and Blood Institute, National Institute of Health, Bethesda, Maryland
| | - J. Rodante
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung and Blood Institute, National Institute of Health, Bethesda, Maryland
| | - A. Keel
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung and Blood Institute, National Institute of Health, Bethesda, Maryland
| | - AV. Sorokin
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung and Blood Institute, National Institute of Health, Bethesda, Maryland
| | - A. Svedbom
- Division of Dermatology and Venerology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - HL. Teague
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung and Blood Institute, National Institute of Health, Bethesda, Maryland
| | - M. Stahle
- Division of Dermatology and Venerology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - NN. Mehta
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung and Blood Institute, National Institute of Health, Bethesda, Maryland
| | - MP. Playford
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung and Blood Institute, National Institute of Health, Bethesda, Maryland
| |
Collapse
|
35
|
Tittes J, Brell J, Fritz P, Jonak C, Stary G, Ressler JM, Künig S, Weninger W, Stöckl J. Regulation of the Immune Cell Repertoire in Psoriasis Patients Upon Blockade of IL-17A or TNFα. Dermatol Ther (Heidelb) 2024; 14:613-626. [PMID: 38459237 PMCID: PMC10965886 DOI: 10.1007/s13555-024-01112-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/08/2024] [Indexed: 03/10/2024] Open
Abstract
INTRODUCTION Targeting of the proinflammatory cytokine interleukin 17A (IL-17A) or tumor necrosis factor alpha (TNFα) with the monoclonal antibodies (mAbs) ixekizumab or adalimumab, respectively, is a successful therapy for chronic plaque psoriasis. The effects of these treatments on immune cell populations in the skin are largely unknown. METHODS In this study, we compared the composition of cutaneous, lesional and non-lesional immune cells and blood immune cells in ixekizumab- or adalimumab-treated patients with psoriasis. RESULTS Our data reveal that both treatments efficiently downregulate T cells, macrophages and different subsets of dendritic cells (DCs) in lesional skin towards levels of healthy skin. In contrast to lesional skin, non-lesional areas in patients harbor only few or no detectable DCs compared to the skin of healthy subjects. Treatment with neither ixekizumab nor adalimumab reversed this DC imbalance in non-lesional skin of psoriatic patients. CONCLUSION Our study shows that anti-IL-17A and anti-TNFα therapy rebalances the immune cell repertoire of lesional skin in psoriatic patients but fails to restore the disturbed immune cell repertoire in non-lesional skin.
Collapse
Affiliation(s)
- Julia Tittes
- Department of Dermatology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| | - Jennifer Brell
- Institute of Immunology, Medical University of Vienna, Lazarettgasse 19, 1090, Vienna, Austria
| | - Pia Fritz
- Institute of Immunology, Medical University of Vienna, Lazarettgasse 19, 1090, Vienna, Austria
| | - Constanze Jonak
- Department of Dermatology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Georg Stary
- Department of Dermatology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Julia M Ressler
- Department of Dermatology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Sarojinidevi Künig
- Institute of Immunology, Medical University of Vienna, Lazarettgasse 19, 1090, Vienna, Austria
| | - Wolfgang Weninger
- Department of Dermatology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Johannes Stöckl
- Institute of Immunology, Medical University of Vienna, Lazarettgasse 19, 1090, Vienna, Austria
| |
Collapse
|
36
|
Benezeder T, Bordag N, Woltsche J, Teufelberger A, Perchthaler I, Weger W, Salmhofer W, Gruber-Wackernagel A, Painsi C, Zhan Q, El-Heliebi A, Babina M, Clark R, Wolf P. Mast cells express IL17A, IL17F and RORC, are activated and persist with IL-17 production in resolved skin of patients with chronic plaque-type psoriasis. RESEARCH SQUARE 2024:rs.3.rs-3958361. [PMID: 38410434 PMCID: PMC10896398 DOI: 10.21203/rs.3.rs-3958361/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Little is known about IL-17 expression in psoriasis and the actual cellular source of IL-17 remains incompletely defined. We show that high numbers of IL-17 + mast cells persisted in resolved lesions after treatment (anti-IL-17A, anti-IL-23, UVB or topical dithranol) and correlated inversely with the time span in remission. IL-17 + mast cells were found in T cell-rich areas and often close to resident memory T cells (Trm) in active psoriasis and resolved lesional skin. Digital cytometry by deconvolution of RNA-seq data showed that activated mast cells were increased in psoriatic skin, while resting mast cells were almost absent and both returned to normal levels after treatment. When primary human skin mast cells were stimulated with T cell cytokines (TNFα, IL-22 and IFNγ), they responded by releasing more IL-17A, as measured by ELISA. In situ mRNA detection using padlock probes specific for transcript variants of IL17A, IL17F, and RORC (encoding the Th17 transcription factor RORγt) revealed positive mRNA signals for IL17A, IL17F, and RORCin tryptase + cells, demonstrating that mast cells have the transcriptional machinery to actively produce IL-17. Mast cells thus belong to the center of the IL-23/IL-17 axis and high numbers of IL-17 + mast cells predict an earlier disease recurrence.
Collapse
Affiliation(s)
- Theresa Benezeder
- Department of Dermatology and Venereology, Medical University of Graz
| | - Natalie Bordag
- Department of Dermatology and Venereology, Medical University of Graz
| | - Johannes Woltsche
- Department of Dermatology and Venereology, Medical University of Graz
| | | | | | - Wolfgang Weger
- Department of Dermatology and Venereology, Medical University of Graz
| | | | | | | | - Qian Zhan
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School
| | - Amin El-Heliebi
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz
| | - Magda Babina
- Institute of Allergology, Charite-Universitatsmedizin Berlin
| | | | - Peter Wolf
- Department of Dermatology and Venereology, Medical University of Graz, Graz, Austria
| |
Collapse
|
37
|
Bissonnette R, Pinter A, Ferris LK, Gerdes S, Rich P, Vender R, Miller M, Shen YK, Kannan A, Li S, DeKlotz C, Papp K. An Oral Interleukin-23-Receptor Antagonist Peptide for Plaque Psoriasis. N Engl J Med 2024; 390:510-521. [PMID: 38324484 DOI: 10.1056/nejmoa2308713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
BACKGROUND The use of monoclonal antibodies has changed the treatment of several immune-mediated inflammatory diseases, including psoriasis. However, these large proteins must be administered by injection. JNJ-77242113 is a novel, orally administered interleukin-23-receptor antagonist peptide that selectively blocks interleukin-23 signaling and downstream cytokine production. METHODS In this phase 2 dose-finding trial, we randomly assigned patients with moderate-to-severe plaque psoriasis to receive JNJ-77242113 at a dose of 25 mg once daily, 25 mg twice daily, 50 mg once daily, 100 mg once daily, or 100 mg twice daily or placebo for 16 weeks. The primary end point was a reduction from baseline of at least 75% in the Psoriasis Area and Severity Index (PASI) score (PASI 75 response; PASI scores range from 0 to 72, with higher scores indicating greater extent or severity of psoriasis) at week 16. RESULTS A total of 255 patients underwent randomization. The mean PASI score at baseline was 19.1. The mean duration of psoriasis was 18.2 years, and 78% of the patients across all the trial groups had previously received systemic treatments. At week 16, the percentages of patients with a PASI 75 response were higher among those in the JNJ-77242113 groups (37%, 51%, 58%, 65%, and 79% in the 25-mg once-daily, 25-mg twice-daily, 50-mg once-daily, 100-mg once-daily, and 100-mg twice-daily groups, respectively) than among those in the placebo group (9%), a finding that showed a significant dose-response relationship (P<0.001). The most common adverse events included coronavirus disease 2019 (in 12% of the patients in the placebo group and in 11% of those across the JNJ-77242113 dose groups) and nasopharyngitis (in 5% and 7%, respectively). The percentages of patients who had at least one adverse event were similar in the combined JNJ-77242113 dose group (52%) and the placebo group (51%). There was no evidence of a dose-related increase in adverse events across the JNJ-77242113 dose groups. CONCLUSIONS After 16 weeks of once- or twice-daily oral administration, treatment with the interleukin-23-receptor antagonist peptide JNJ-77242113 showed greater efficacy than placebo in patients with moderate-to-severe plaque psoriasis. (Funded by Janssen Research and Development; FRONTIER 1 ClinicalTrials.gov number, NCT05223868.).
Collapse
Affiliation(s)
- Robert Bissonnette
- From Innovaderm Research, Montreal (R.B.), McMaster University and Dermatrials Research, Hamilton, ON (R.V.), Alliance Clinical Trials and Probity Medical Research, Waterloo, ON (K.P.), and the Division of Dermatology, Department of Medicine, University of Toronto, Toronto (K.P.) - all in Canada; Goethe University Frankfurt, Frankfurt am Main (A.P.), and the Center for Inflammatory Skin Diseases, University Medical Center Schleswig-Holstein Campus Kiel, Kiel (S.G.) - both in Germany; the University of Pittsburgh, Pittsburgh (L.K.F.), and Janssen Research and Development, Spring House (M.M., Y.-K.S., A.K., S.L., C.D.) - both in Pennsylvania; and Oregon Dermatology and Research Center, Portland (P.R.)
| | - Andreas Pinter
- From Innovaderm Research, Montreal (R.B.), McMaster University and Dermatrials Research, Hamilton, ON (R.V.), Alliance Clinical Trials and Probity Medical Research, Waterloo, ON (K.P.), and the Division of Dermatology, Department of Medicine, University of Toronto, Toronto (K.P.) - all in Canada; Goethe University Frankfurt, Frankfurt am Main (A.P.), and the Center for Inflammatory Skin Diseases, University Medical Center Schleswig-Holstein Campus Kiel, Kiel (S.G.) - both in Germany; the University of Pittsburgh, Pittsburgh (L.K.F.), and Janssen Research and Development, Spring House (M.M., Y.-K.S., A.K., S.L., C.D.) - both in Pennsylvania; and Oregon Dermatology and Research Center, Portland (P.R.)
| | - Laura K Ferris
- From Innovaderm Research, Montreal (R.B.), McMaster University and Dermatrials Research, Hamilton, ON (R.V.), Alliance Clinical Trials and Probity Medical Research, Waterloo, ON (K.P.), and the Division of Dermatology, Department of Medicine, University of Toronto, Toronto (K.P.) - all in Canada; Goethe University Frankfurt, Frankfurt am Main (A.P.), and the Center for Inflammatory Skin Diseases, University Medical Center Schleswig-Holstein Campus Kiel, Kiel (S.G.) - both in Germany; the University of Pittsburgh, Pittsburgh (L.K.F.), and Janssen Research and Development, Spring House (M.M., Y.-K.S., A.K., S.L., C.D.) - both in Pennsylvania; and Oregon Dermatology and Research Center, Portland (P.R.)
| | - Sascha Gerdes
- From Innovaderm Research, Montreal (R.B.), McMaster University and Dermatrials Research, Hamilton, ON (R.V.), Alliance Clinical Trials and Probity Medical Research, Waterloo, ON (K.P.), and the Division of Dermatology, Department of Medicine, University of Toronto, Toronto (K.P.) - all in Canada; Goethe University Frankfurt, Frankfurt am Main (A.P.), and the Center for Inflammatory Skin Diseases, University Medical Center Schleswig-Holstein Campus Kiel, Kiel (S.G.) - both in Germany; the University of Pittsburgh, Pittsburgh (L.K.F.), and Janssen Research and Development, Spring House (M.M., Y.-K.S., A.K., S.L., C.D.) - both in Pennsylvania; and Oregon Dermatology and Research Center, Portland (P.R.)
| | - Phoebe Rich
- From Innovaderm Research, Montreal (R.B.), McMaster University and Dermatrials Research, Hamilton, ON (R.V.), Alliance Clinical Trials and Probity Medical Research, Waterloo, ON (K.P.), and the Division of Dermatology, Department of Medicine, University of Toronto, Toronto (K.P.) - all in Canada; Goethe University Frankfurt, Frankfurt am Main (A.P.), and the Center for Inflammatory Skin Diseases, University Medical Center Schleswig-Holstein Campus Kiel, Kiel (S.G.) - both in Germany; the University of Pittsburgh, Pittsburgh (L.K.F.), and Janssen Research and Development, Spring House (M.M., Y.-K.S., A.K., S.L., C.D.) - both in Pennsylvania; and Oregon Dermatology and Research Center, Portland (P.R.)
| | - Ronald Vender
- From Innovaderm Research, Montreal (R.B.), McMaster University and Dermatrials Research, Hamilton, ON (R.V.), Alliance Clinical Trials and Probity Medical Research, Waterloo, ON (K.P.), and the Division of Dermatology, Department of Medicine, University of Toronto, Toronto (K.P.) - all in Canada; Goethe University Frankfurt, Frankfurt am Main (A.P.), and the Center for Inflammatory Skin Diseases, University Medical Center Schleswig-Holstein Campus Kiel, Kiel (S.G.) - both in Germany; the University of Pittsburgh, Pittsburgh (L.K.F.), and Janssen Research and Development, Spring House (M.M., Y.-K.S., A.K., S.L., C.D.) - both in Pennsylvania; and Oregon Dermatology and Research Center, Portland (P.R.)
| | - Megan Miller
- From Innovaderm Research, Montreal (R.B.), McMaster University and Dermatrials Research, Hamilton, ON (R.V.), Alliance Clinical Trials and Probity Medical Research, Waterloo, ON (K.P.), and the Division of Dermatology, Department of Medicine, University of Toronto, Toronto (K.P.) - all in Canada; Goethe University Frankfurt, Frankfurt am Main (A.P.), and the Center for Inflammatory Skin Diseases, University Medical Center Schleswig-Holstein Campus Kiel, Kiel (S.G.) - both in Germany; the University of Pittsburgh, Pittsburgh (L.K.F.), and Janssen Research and Development, Spring House (M.M., Y.-K.S., A.K., S.L., C.D.) - both in Pennsylvania; and Oregon Dermatology and Research Center, Portland (P.R.)
| | - Yaung-Kaung Shen
- From Innovaderm Research, Montreal (R.B.), McMaster University and Dermatrials Research, Hamilton, ON (R.V.), Alliance Clinical Trials and Probity Medical Research, Waterloo, ON (K.P.), and the Division of Dermatology, Department of Medicine, University of Toronto, Toronto (K.P.) - all in Canada; Goethe University Frankfurt, Frankfurt am Main (A.P.), and the Center for Inflammatory Skin Diseases, University Medical Center Schleswig-Holstein Campus Kiel, Kiel (S.G.) - both in Germany; the University of Pittsburgh, Pittsburgh (L.K.F.), and Janssen Research and Development, Spring House (M.M., Y.-K.S., A.K., S.L., C.D.) - both in Pennsylvania; and Oregon Dermatology and Research Center, Portland (P.R.)
| | - Arun Kannan
- From Innovaderm Research, Montreal (R.B.), McMaster University and Dermatrials Research, Hamilton, ON (R.V.), Alliance Clinical Trials and Probity Medical Research, Waterloo, ON (K.P.), and the Division of Dermatology, Department of Medicine, University of Toronto, Toronto (K.P.) - all in Canada; Goethe University Frankfurt, Frankfurt am Main (A.P.), and the Center for Inflammatory Skin Diseases, University Medical Center Schleswig-Holstein Campus Kiel, Kiel (S.G.) - both in Germany; the University of Pittsburgh, Pittsburgh (L.K.F.), and Janssen Research and Development, Spring House (M.M., Y.-K.S., A.K., S.L., C.D.) - both in Pennsylvania; and Oregon Dermatology and Research Center, Portland (P.R.)
| | - Shu Li
- From Innovaderm Research, Montreal (R.B.), McMaster University and Dermatrials Research, Hamilton, ON (R.V.), Alliance Clinical Trials and Probity Medical Research, Waterloo, ON (K.P.), and the Division of Dermatology, Department of Medicine, University of Toronto, Toronto (K.P.) - all in Canada; Goethe University Frankfurt, Frankfurt am Main (A.P.), and the Center for Inflammatory Skin Diseases, University Medical Center Schleswig-Holstein Campus Kiel, Kiel (S.G.) - both in Germany; the University of Pittsburgh, Pittsburgh (L.K.F.), and Janssen Research and Development, Spring House (M.M., Y.-K.S., A.K., S.L., C.D.) - both in Pennsylvania; and Oregon Dermatology and Research Center, Portland (P.R.)
| | - Cynthia DeKlotz
- From Innovaderm Research, Montreal (R.B.), McMaster University and Dermatrials Research, Hamilton, ON (R.V.), Alliance Clinical Trials and Probity Medical Research, Waterloo, ON (K.P.), and the Division of Dermatology, Department of Medicine, University of Toronto, Toronto (K.P.) - all in Canada; Goethe University Frankfurt, Frankfurt am Main (A.P.), and the Center for Inflammatory Skin Diseases, University Medical Center Schleswig-Holstein Campus Kiel, Kiel (S.G.) - both in Germany; the University of Pittsburgh, Pittsburgh (L.K.F.), and Janssen Research and Development, Spring House (M.M., Y.-K.S., A.K., S.L., C.D.) - both in Pennsylvania; and Oregon Dermatology and Research Center, Portland (P.R.)
| | - Kim Papp
- From Innovaderm Research, Montreal (R.B.), McMaster University and Dermatrials Research, Hamilton, ON (R.V.), Alliance Clinical Trials and Probity Medical Research, Waterloo, ON (K.P.), and the Division of Dermatology, Department of Medicine, University of Toronto, Toronto (K.P.) - all in Canada; Goethe University Frankfurt, Frankfurt am Main (A.P.), and the Center for Inflammatory Skin Diseases, University Medical Center Schleswig-Holstein Campus Kiel, Kiel (S.G.) - both in Germany; the University of Pittsburgh, Pittsburgh (L.K.F.), and Janssen Research and Development, Spring House (M.M., Y.-K.S., A.K., S.L., C.D.) - both in Pennsylvania; and Oregon Dermatology and Research Center, Portland (P.R.)
| |
Collapse
|
38
|
Alomari N, Alhussaini W. Update on the advances and challenges in bioequivalence testing methods for complex topical generic products. Front Pharmacol 2024; 15:1330712. [PMID: 38389924 PMCID: PMC10881717 DOI: 10.3389/fphar.2024.1330712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
Most of the government regulatory agencies, including the United States Food and Drug Administration and the European Medicine Agency, demand that the generic complex topical products prove pharmaceutical and bioequivalence. The evaluation of bioequivalence for complex topical dermatological formulations is a challenging task that requires careful consideration of several factors. Although comparative clinical studies are still considered the gold standard approach for establishing bioequivalence in most formulations, these studies can be costly and insensitive to detect formulation differences. Therefore, significant efforts have been made to develop and validate alternative approaches that demonstrate bioequivalence and expedite the availability of high-quality generic topical dermatological products. This article reviews the current methods for determining the bioequivalence of topical formulations in humans, with particular emphasis on recent advances in these methodologies. Most of the alternative methods are sensitive and reproducible, with the capability to ease the financial burden of comparative clinical studies within a short delivery time. The limitations associated with each technique are reviewed in detail.
Collapse
Affiliation(s)
- Nedaa Alomari
- Department of Pharmaceutical Analysis, King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, Ministry of the National Guard-Health Affairs, Riyadh, Saudi Arabia
| | - Waleed Alhussaini
- Department of Pharmaceutical Analysis, King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, Ministry of the National Guard-Health Affairs, Riyadh, Saudi Arabia
| |
Collapse
|
39
|
Tanaka Y, Shaw S. Bimekizumab for the treatment of psoriatic arthritis. Expert Rev Clin Immunol 2024; 20:155-168. [PMID: 37909894 DOI: 10.1080/1744666x.2023.2277266] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/26/2023] [Indexed: 11/03/2023]
Abstract
INTRODUCTION Interleukin (IL)-17A and IL-17F have overlapping roles in pro-inflammatory signaling and have been implicated in the pathogenesis of psoriatic disease. Bimekizumab is the first human monoclonal antibody to selectively inhibit IL-17F in addition to IL-17A. Bimekizumab has been studied in several phase II/III trials and has been approved for the treatment of patients with psoriatic arthritis (PsA) in the EU and UK. AREAS COVERED A literature search identified clinical trials examining the efficacy and safety of bimekizumab for PsA, which were critically appraised. EXPERT OPINION Clinical trials of bimekizumab in PsA have demonstrated rapid and sustained treatment responses and depth of response across the multiple disease domains. High levels of efficacy were sustained to 152 weeks in phase IIb trials, and to 52 weeks in phase III trials. Bimekizumab was generally well tolerated. As expected, due to the role of IL-17 in the immune response to fungal pathogens, there was an increase in mild-to-moderate, localized fungal infections with bimekizumab treatment, very few of which led to discontinuation. Studies over longer time periods, with relevant comparators from the IL-17A inhibitor class, and real-world data will be important to further define the role of bimekizumab among currently available treatments for PsA. [Figure: see text].
Collapse
Affiliation(s)
- Yoshiya Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| | | |
Collapse
|
40
|
Bodenlenz M, Yeoh T, Berstein G, Mathew S, Shah J, Banfield C, Hollingshead B, Steyn SJ, Osgood SM, Beaumont K, Kainz S, Holeček C, Trausinger G, Raml R, Birngruber T. Comparative Study of Dermal Pharmacokinetics Between Topical Drugs Using Open Flow Microperfusion in a Pig Model. Pharm Res 2024; 41:223-234. [PMID: 38158503 PMCID: PMC10879402 DOI: 10.1007/s11095-023-03645-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 12/14/2023] [Indexed: 01/03/2024]
Abstract
PURPOSE Accurate methods to determine dermal pharmacokinetics are important to increase the rate of clinical success in topical drug development. We investigated in an in vivo pig model whether the unbound drug concentration in the interstitial fluid as determined by dermal open flow microperfusion (dOFM) is a more reliable measure of dermal exposure compared to dermal biopsies for seven prescription or investigational drugs. In addition, we verified standard dOFM measurement using a recirculation approach and compared dosing frequencies (QD versus BID) and dose strengths (high versus low drug concentrations). METHODS Domestic pigs were topically administered seven different drugs twice daily in two studies. On day 7, drug exposures in the dermis were assessed in two ways: (1) dOFM provided the total and unbound drug concentrations in dermal interstitial fluid, and (2) clean punch biopsies after heat separation provided the total concentrations in the upper and lower dermis. RESULTS dOFM showed sufficient intra-study precision to distinguish interstitial fluid concentrations between different drugs, dose frequencies and dose strengths, and had good reproducibility between studies. Biopsy concentrations showed much higher and more variable values. Standard dOFM measurements were consistent with values obtained with the recirculation approach. CONCLUSIONS dOFM pig model is a robust and reproducible method to directly determine topical drug concentration in dermal interstitial fluid. Dermal biopsies were a less reliable measure of dermal exposure due to possible contributions from drug bound to tissue and drug associated with skin appendages.
Collapse
Affiliation(s)
- Manfred Bodenlenz
- HEALTH - Institute for Biomedical Research and Technologies, Joanneum Research Forschungsgesellschaft M.B.H, Neue Stiftingtalstrasse 2, 8010, Graz, Austria
| | - Thean Yeoh
- Pfizer Research Technology Center, 1 Portland St, Cambridge, MA, 02139, USA
| | - Gabriel Berstein
- Pfizer Research Technology Center, 1 Portland St, Cambridge, MA, 02139, USA
| | - Shibin Mathew
- Pfizer Research Technology Center, 1 Portland St, Cambridge, MA, 02139, USA.
| | - Jaymin Shah
- Pfizer Research Technology Center, 1 Portland St, Cambridge, MA, 02139, USA
| | | | - Brett Hollingshead
- Pfizer Research Technology Center, 1 Portland St, Cambridge, MA, 02139, USA
| | - Stefanus J Steyn
- Pfizer Research Technology Center, 1 Portland St, Cambridge, MA, 02139, USA
| | - Sarah M Osgood
- Pfizer Research Technology Center, 1 Portland St, Cambridge, MA, 02139, USA
| | - Kevin Beaumont
- Pfizer Research Technology Center, 1 Portland St, Cambridge, MA, 02139, USA
| | - Sonja Kainz
- HEALTH - Institute for Biomedical Research and Technologies, Joanneum Research Forschungsgesellschaft M.B.H, Neue Stiftingtalstrasse 2, 8010, Graz, Austria
| | - Christian Holeček
- HEALTH - Institute for Biomedical Research and Technologies, Joanneum Research Forschungsgesellschaft M.B.H, Neue Stiftingtalstrasse 2, 8010, Graz, Austria
| | - Gert Trausinger
- HEALTH - Institute for Biomedical Research and Technologies, Joanneum Research Forschungsgesellschaft M.B.H, Neue Stiftingtalstrasse 2, 8010, Graz, Austria
| | - Reingard Raml
- HEALTH - Institute for Biomedical Research and Technologies, Joanneum Research Forschungsgesellschaft M.B.H, Neue Stiftingtalstrasse 2, 8010, Graz, Austria
| | - Thomas Birngruber
- HEALTH - Institute for Biomedical Research and Technologies, Joanneum Research Forschungsgesellschaft M.B.H, Neue Stiftingtalstrasse 2, 8010, Graz, Austria.
| |
Collapse
|
41
|
Quah S, Sundaram GM, Subramanian G, Vaz C, Tan JSL, Kabir RF, Ong JMR, Oon HH, Theng C, Sampath P. IL-17-Mediated Downregulation of miR-101 Facilitates the Expression of EZH2 to Promote Epidermal Hyperplasia in Psoriasis. J Invest Dermatol 2024; 144:403-407.e7. [PMID: 37574185 DOI: 10.1016/j.jid.2023.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 08/15/2023]
Affiliation(s)
- Shan Quah
- A∗STAR Skin Research Labs (A∗SRL), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | | | - Gowtham Subramanian
- A∗STAR Skin Research Labs (A∗SRL), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Candida Vaz
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Jonathan S L Tan
- A∗STAR Skin Research Labs (A∗SRL), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Ramisa Fariha Kabir
- A∗STAR Skin Research Labs (A∗SRL), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Jesslyn M R Ong
- A∗STAR Skin Research Labs (A∗SRL), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Hazel H Oon
- Skin Research Institute of Singapore (SRIS), Singapore, Singapore; Division of Dermatology, National Skin Centre, Singapore, Singapore
| | - Colin Theng
- The Skin Specialist & Laser Clinic, Mount Alvernia Medical Centre, Singapore, Singapore
| | - Prabha Sampath
- A∗STAR Skin Research Labs (A∗SRL), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Skin Research Institute of Singapore (SRIS), Singapore, Singapore; Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Program in Cancer & Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
42
|
Rusiñol L, Puig L. Multi-Omics Approach to Improved Diagnosis and Treatment of Atopic Dermatitis and Psoriasis. Int J Mol Sci 2024; 25:1042. [PMID: 38256115 PMCID: PMC10815999 DOI: 10.3390/ijms25021042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 01/07/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Psoriasis and atopic dermatitis fall within the category of cutaneous immune-mediated inflammatory diseases (IMIDs). The prevalence of IMIDs is increasing in industrialized societies, influenced by both environmental changes and a genetic predisposition. However, the exact immune factors driving these chronic, progressive diseases are not fully understood. By using multi-omics techniques in cutaneous IMIDs, it is expected to advance the understanding of skin biology, uncover the underlying mechanisms of skin conditions, and potentially devise precise and personalized approaches to diagnosis and treatment. We provide a narrative review of the current knowledge in genomics, epigenomics, and proteomics of atopic dermatitis and psoriasis. A literature search was performed for articles published until 30 November 2023. Although there is still much to uncover, recent evidence has already provided valuable insights, such as proteomic profiles that permit differentiating psoriasis from mycosis fungoides and β-defensin 2 correlation to PASI and its drop due to secukinumab first injection, among others.
Collapse
Affiliation(s)
- Lluís Rusiñol
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
- Institut de Recerca Sant Pau (IR SANT PAU), 08041 Barcelona, Spain
- Unitat Docent Hospital Universitari Sant Pau, Universitat Autònoma de Barcelona, 08025 Barcelona, Spain
| | - Lluís Puig
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
- Institut de Recerca Sant Pau (IR SANT PAU), 08041 Barcelona, Spain
- Unitat Docent Hospital Universitari Sant Pau, Universitat Autònoma de Barcelona, 08025 Barcelona, Spain
| |
Collapse
|
43
|
Qi F, Jin H. Extracellular vesicles from keratinocytes and other skin-related cells in psoriasis: A review. Exp Dermatol 2024; 33:e15001. [PMID: 38284192 DOI: 10.1111/exd.15001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 11/13/2023] [Accepted: 11/25/2023] [Indexed: 01/30/2024]
Abstract
Psoriasis is a highly prevalent chronic inflammatory skin condition involving abnormal proliferation and differentiation of keratinocytes, together with substantial infiltration of immune cells. Extracellular vesicles (EVs), which are released spontaneously into the extracellular space by virtually all cell types, play a crucial role in cell-to-cell communication by delivering bioactive cargos such as mRNA nucleic acids and proteins to recipient cells. Numerous studies have highlighted the significant contributions of EVs to both the pathogenesis and treatment of psoriasis. This review provides a concise overview of skin-derived EVs and their involvement in the pathogenesis of psoriasis.
Collapse
Affiliation(s)
- Fei Qi
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Hongzhong Jin
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| |
Collapse
|
44
|
Matsuno A, Sumida H, Nakanishi H, Ikeyama Y, Ishii T, Omori I, Saito H, Iwasawa O, Sugimori A, Yoshizaki A, Katoh H, Ishikawa S, Sato S. Keratinocyte proline-rich protein modulates immune and epidermal response in imiquimod-induced psoriatic skin inflammation. Exp Dermatol 2023; 32:2121-2130. [PMID: 37926955 DOI: 10.1111/exd.14960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/28/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023]
Abstract
Psoriasis is a persistent inflammatory skin disease thought to arise as a result of the infiltration of inflammatory cells and activation of keratinocytes. Recent advances in basic research and clinical experience revealed that the interleukin (IL)-23/IL-17 axis has been identified as a major immune pathway in psoriasis. However, it remains unclear how keratinocyte factors contribute to the pathology of psoriasis. Keratinocyte proline-rich protein (KPRP) is a proline-rich insoluble protein, which is present in the epidermis and is likely to be involved in the skin barrier function. Here, to investigate the potential roles of KPRP in psoriatic skin inflammation, Kprp-modified mice were applied in the imiquimod (IMQ)-induced skin inflammation model, which develops psoriasis-like epidermal hyperplasia and cutaneous inflammation features. Then, heterozygous knockout (Kprp+/- ) but not homozygous knockout (Kprp-/- ) mice displayed attenuated skin erythema compared to control wild-type mice. In addition, RNA sequencing, quantitative PCR and/or histological analysis detected changes in the expression of several molecules related to psoriatic inflammation or keratinocyte differentiation in Kprp+/- mice, but not Kprp-/- mice. Further analysis exhibited reduced IL-17-producing γδlow T cells and amplified epidermal hyperplasia in Kprp+/- mice, which were implied to be related to decreased expression of β-defensins and increased expression of LPAR1 (Lysophosphatidic acid receptor 1), respectively. Thus, our results imply that KPRP has the potential as a therapeutic target in psoriatic skin inflammation.
Collapse
Affiliation(s)
- Ai Matsuno
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hayakazu Sumida
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Scleroderma Center, The University of Tokyo Hospital, Tokyo, Japan
- SLE Center, The University of Tokyo Hospital, Tokyo, Japan
| | - Hirofumi Nakanishi
- Research and Development Division, Rohto Pharmaceutical Company, Osaka, Japan
| | - Yoshifumi Ikeyama
- Research and Development Division, Rohto Pharmaceutical Company, Osaka, Japan
| | - Tsuyoshi Ishii
- Research and Development Division, Rohto Pharmaceutical Company, Osaka, Japan
| | - Issei Omori
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hinako Saito
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Okuto Iwasawa
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ayaka Sugimori
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ayumi Yoshizaki
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroto Katoh
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shumpei Ishikawa
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shinichi Sato
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
45
|
Cui J, Zhang X, Deng J, Yan Y, Yao D, Deng H, Yu J, Ye S, Han L, Yu X, Lu C. Potential biomarkers for psoriasis topical treatment by in-depth serum proteomics. J DERMATOL TREAT 2023; 34:2248318. [PMID: 37621164 DOI: 10.1080/09546634.2023.2248318] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 08/02/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND Psoriasis is a chronic skin disease, and topical sequential therapy with a combination of calcipotriol and calcipotriol betamethasone is currently approved topical treatment. However, the exact mechanism by which this treatment regimen relieves psoriasis is unknown. METHOD We assembled a cohort of 65 psoriasis patients and divided post-treatment cohort into responder group and non-responder group according to the Psoriasis Area Severity Index (PASI) score after 12-week treatment. We measured the expression levels of proteins in collected 130 serum samples using our in-depth proteomics platform with a data-independent acquisition mass spectrometer and antibody microarray. We performed bioinformatics analyses of the biologic processes and signaling pathways that were changed in the responder group and constructed a proteomics landscape of psoriasis pathogenesis response to treatment. We then validated the biomarkers of disease severity in an independent cohort of 88 samples using an enzyme-linked immunosorbent assay. RESULTS We first identified 174 differentially expressed proteins (DEPs) for comparative analysis of proteins between responders and non-responders at baseline (p < 0.05). Then pathway analysis showed that the responders focused more on signaling molecules and interaction, complement and coagulation cascades, whereas the non-responders more on signal transduction and IL-17 signaling pathways. We further identified four candidate biomarkers (COLEC11, C1QA, BNC2, ITIH4) response to treatment. We also found 125 DEPs (p < 0.05) after treatment compared with before treatment in responder group. Pathway analysis showed an enrichment in pathways related to complement and coagulation cascades, phagosome, ECM-receptor interaction, cholesterol metabolism, vitamin digestion and absorption. CD14 was validated as potential biomarkers for the disease severity of psoriasis and treatment targets. CONCLUSION In this work, we analyzed the response to topical sequential therapy and finally identified four biomarkers. Additionally, we found that topical sequential therapy may alleviate psoriasis by regulating lipid metabolism and modulating the immune response by affecting the complement activation process.
Collapse
Affiliation(s)
- Jingwen Cui
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaomei Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Jingwen Deng
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Yuhong Yan
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Danni Yao
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Hao Deng
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Jingjie Yu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Shuyan Ye
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Ling Han
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Xiaobo Yu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Chuanjian Lu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| |
Collapse
|
46
|
Orro K, Salk K, Merkulova A, Abram K, Karelson M, Traks T, Neuman T, Spee P, Kingo K. Non-Invasive Assessment of Skin Surface Proteins of Psoriasis Vulgaris Patients in Response to Biological Therapy. Int J Mol Sci 2023; 24:16248. [PMID: 38003437 PMCID: PMC10671061 DOI: 10.3390/ijms242216248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Measurements of skin surface biomarkers have enormous value for the detailed assessment of skin conditions, both for clinical application and in skin care. The main goals of the current study were to assess whether expression patterns of skin surface hBD-1, hBD-2, IL-1α, CXCL-1, and CXCL-8, examples of proteins known to be involved in psoriasis pathology, are associated with disease severity and whether expression patterns of these proteins on the skin surface can be used to measure pharmacodynamic effects of biological therapy. In this observational study using transdermal analysis patch (TAP), levels of skin surface IL-1α, hBD-1, hBD-2, CXCL-1/2, and CXCL-8 of psoriasis vulgaris (PV) patients over biological therapy were assessed. The Psoriasis Area Severity Index (PASI) and local score for erythema, induration, and desquamation were determined from the exact same skin area as FibroTx TAP measurements. Thirty-seven adult PV patients were included, of which twenty-three were subjected to anti-TNF-α, seven to anti-IL-17A, and seven to anti-IL12/IL-23 therapy. Significantly higher levels of hBD-1, hBD-2, CXCL-1/2, and CXCL-8 were detected on lesional skin compared to the non-lesional skin of the PV patients. In contrast, lower levels of IL-1α were found in lesional skin compared to non-lesional skin. In addition, we observed that the biomarker expression levels correlate with disease severity. Further, we confirmed that changes in the expression levels of skin surface biomarkers during biological therapy correlate with treatment response. Biomarker expression patterns in response to treatment differed somewhat between treatment subtypes. We observed that, in the case of anti-TNF-α therapy, an increase after a steady decrease in the expression levels of CXCL-1/2 and CXCL-8 occurred before the change in clinical scores. Moreover, response kinetics of skin surface proteins differs between the applied therapies-hBD2 expression responds quickly to anti-IL-17A therapy, CXCL-1/2 to anti-IL-12/23, and levels of CXCL-8 are rapidly down-regulated by IL-17A and IL-12/23 therapy. Our findings confirm that the skin surface hBD-2, IL-1α, CXCL-1/2, and CXCL-8 are markers for the psoriasis severity. Further, data obtained during this study give the basis for the conclusion that skin surface proteins CXCL-1/2 and CXCL-8 may have value as therapeutic biomarkers, thus confirming that measuring the 'molecular root' of inflammation appears to have value in scoring disease severity on its own.
Collapse
Affiliation(s)
- Kadri Orro
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee, 12618 Tallinn, Estonia;
- FibroTx LLC., Mäealuse 4, 12618 Tallinn, Estonia (A.M.); (P.S.)
| | - Kristiina Salk
- FibroTx LLC., Mäealuse 4, 12618 Tallinn, Estonia (A.M.); (P.S.)
| | - Anna Merkulova
- FibroTx LLC., Mäealuse 4, 12618 Tallinn, Estonia (A.M.); (P.S.)
| | - Kristi Abram
- Clinic of Dermatology, Tartu University Hospital, 50417 Tartu, Estonia
- Clinic of Dermatology, Institute of Clinical Medicine, Tartu University, 50417 Tartu, Estonia
| | - Maire Karelson
- Clinic of Dermatology, Tartu University Hospital, 50417 Tartu, Estonia
- Clinic of Dermatology, Institute of Clinical Medicine, Tartu University, 50417 Tartu, Estonia
| | - Tanel Traks
- Clinic of Dermatology, Institute of Clinical Medicine, Tartu University, 50417 Tartu, Estonia
| | - Toomas Neuman
- FibroTx LLC., Mäealuse 4, 12618 Tallinn, Estonia (A.M.); (P.S.)
| | - Pieter Spee
- FibroTx LLC., Mäealuse 4, 12618 Tallinn, Estonia (A.M.); (P.S.)
- PS! Pharmaconsult, Moellemoseparken 44, 3450 Alleroed, Denmark
| | - Külli Kingo
- Clinic of Dermatology, Tartu University Hospital, 50417 Tartu, Estonia
- Clinic of Dermatology, Institute of Clinical Medicine, Tartu University, 50417 Tartu, Estonia
| |
Collapse
|
47
|
Tout I, Noack M, Miossec P. Differential effects of interleukin-17A and 17F on cell interactions between immune cells and stromal cells from synovium or skin. Sci Rep 2023; 13:19223. [PMID: 37932356 PMCID: PMC10628108 DOI: 10.1038/s41598-023-45653-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 10/22/2023] [Indexed: 11/08/2023] Open
Abstract
We compared the contribution of IL-17A and IL-17F in co-culture systems mimicking cell interactions as found in inflamed synovium and skin. Synoviocytes or skin fibroblasts were co-cultured with activated PBMC, with IL-17A, IL-17 A/F, IL-17F, IL-23, anti-IL-17A, anti-IL-17A/F or anti-IL-17F antibodies. IL-17A, IL-17F, IL-6 and IL-10 production was measured at 48 h. mRNA expression of receptor subunits for IL-23, IL-12 and IL-17 was assessed at 24 h. Both cell activation and interactions were needed for a high IL-17A secretion while IL-17F was stimulated by PHA activation alone and further increased in co-cultures. IL-17F levels were higher than IL-17A in both co-cultures (p < 0.05). IL-17F addition decreased IL-17A secretion (p < 0.05) but IL-17A addition had no effect on IL-17F secretion. Interestingly, IL-17A and IL-17F upregulated IL-17RA and IL-17RC mRNA expression in PBMC/skin fibroblast co-cultures (p < 0.05) while only IL-17F exerted this effect in synoviocytes (p < 0.05). Monocyte exclusion in both co-cultures increased IL-17A and IL-17F (twofold, p < 0.05) while decreasing IL-10 and IL-6 secretion (twofold, p < 0.05). IL-17A and F had differential effects on their receptor expression with a higher sensitivity for skin fibroblasts highlighting the differential contribution of IL-17A and F in joint vs. skin diseases.
Collapse
Affiliation(s)
- Issam Tout
- Immunogenomics and Inflammation Research Unit, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 Place d'Arsonval, 69003, Lyon, France
| | - Mélissa Noack
- Immunogenomics and Inflammation Research Unit, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 Place d'Arsonval, 69003, Lyon, France
| | - Pierre Miossec
- Immunogenomics and Inflammation Research Unit, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 Place d'Arsonval, 69003, Lyon, France.
- Department of Clinical Immunology and Rheumatology, Edouard Herriot Hospital, 5 Place d'Arsonval, 69437, Lyon, France.
| |
Collapse
|
48
|
Hurme P, Sahla R, Rückert B, Vahlberg T, Turunen R, Vuorinen T, Akdis M, Söderlund‐Venermo M, Akdis C, Jartti T. Human bocavirus 1 coinfection is associated with decreased cytokine expression in the rhinovirus-induced first wheezing episode in children. Clin Transl Allergy 2023; 13:e12311. [PMID: 38006383 PMCID: PMC10642552 DOI: 10.1002/clt2.12311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/20/2023] [Accepted: 10/22/2023] [Indexed: 11/27/2023] Open
Abstract
BACKGROUND Rhinovirus (RV)-induced first wheezing episodes in children are associated with a markedly increased risk of asthma. Previous studies have suggested that human bocavirus 1 (HBoV1) may modify RV-induced immune responses in young children. We investigated cytokine profiles of sole RV- and dual RV-HBoV1-induced first wheezing episodes, and their association with severity and prognosis. METHODS Fifty-two children infected with only RV and nine children infected with dual RV-HBoV1, aged 3-23 months, with severe first wheezing episodes were recruited. At acute illness and 2 weeks later, peripheral blood mononuclear cells were isolated, and stimulated with anti-CD3/anti-CD28 in vitro. Multiplex ELISA was used to quantitatively identify 56 different cytokines at both study points. Patients were prospectively followed for 4 years. RESULTS The mean age of the children was 14.3 months, and 30% were sensitized. During the acute illness, the adjusted analyses revealed a decrease in the expression of IL-1b, MIP-1b, Regulated upon activation, normal T cell expressed and presumably secreted (CCL5), TNF-a, TARC, and ENA-78 in the RV-HBoV1 group compared with the RV group. In the convalescence phase, the RV-HBoV1 group was characterized by decreased expression of Fractalkine, MCP-3, and IL-8 compared to the RV group. Furthermore, the hospitalization time was associated with the virus group and cytokine response (interaction p < 0.05), signifying that increased levels of epidermal growth factor and MIP-1b were related with a shorter duration of hospitalization in the RV-HBoV1 coinfection group but not in the RV group. CONCLUSIONS Different cytokine response profiles were detected between the RV and the RV-HBoV1 groups. Our results show the idea that RV-induced immune responses may be suppressed by HBoV1.
Collapse
Affiliation(s)
- Pekka Hurme
- Department of Pediatrics and Adolescent MedicineTurku University HospitalUniversity of TurkuTurkuFinland
| | - Reetta Sahla
- Department of Pediatrics and Adolescent MedicineTurku University HospitalUniversity of TurkuTurkuFinland
| | - Beate Rückert
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZürichChristine Kühne‐Center for Allergy Research and Education (CK‐CARE)DavosSwitzerland
| | - Tero Vahlberg
- Department of BiostatisticsUniversity of TurkuTurkuFinland
| | - Riitta Turunen
- Department of Pediatrics and Adolescent MedicineTurku University HospitalUniversity of TurkuTurkuFinland
- New Children's HospitalHelsinki University HospitalUniversity of HelsinkiHelsinkiFinland
| | - Tytti Vuorinen
- Institute of BiomedicineUniversity of TurkuTurkuFinland
- Department of Clinical MicrobiologyTurku University HospitalTurkuFinland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZürichChristine Kühne‐Center for Allergy Research and Education (CK‐CARE)DavosSwitzerland
| | | | - Cezmi Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZürichChristine Kühne‐Center for Allergy Research and Education (CK‐CARE)DavosSwitzerland
| | - Tuomas Jartti
- Department of Pediatrics and Adolescent MedicineTurku University HospitalUniversity of TurkuTurkuFinland
- Research Unit of Clinical MedicineMedical Research CenterUniversity of OuluOuluFinland
- Department of Pediatrics and Adolescent MedicineOulu University HospitalOuluFinland
| |
Collapse
|
49
|
Schäkel K, Reich K, Asadullah K, Pinter A, Jullien D, Weisenseel P, Paul C, Gomez M, Wegner S, Personke Y, Kreimendahl F, Chen Y, Angsana J, Leung MWL, Eyerich K. Early disease intervention with guselkumab in psoriasis leads to a higher rate of stable complete skin clearance ('clinical super response'): Week 28 results from the ongoing phase IIIb randomized, double-blind, parallel-group, GUIDE study. J Eur Acad Dermatol Venereol 2023; 37:2016-2027. [PMID: 37262309 DOI: 10.1111/jdv.19236] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 04/25/2023] [Indexed: 06/03/2023]
Abstract
BACKGROUND Guselkumab is an interleukin (IL)-23 inhibitor with demonstrated efficacy in patients with psoriasis. OBJECTIVES Evaluate the impact of early disease intervention on clinical responses following 28 weeks of guselkumab treatment in patients with moderate-to-severe plaque psoriasis. Correlate clinical response and disease duration data with serum biomarker data. METHODS GUIDE is a phase IIIb randomized, double-blind, parallel-group, multicentre study of adults with moderate-to-severe plaque psoriasis. In study part 1, patients with a short disease duration (SDD [≤2 years]) or a long disease duration (LDD [>2 years]) received guselkumab 100 mg at Week (W) 0, 4, 12, and 20. Those achieving complete skin clearance at W20 and W28 were defined as a super responder (SRe). A multivariable logistic regression analysed the association between baseline factors and the likelihood of becoming an SRe. The relationship between clinical response, disease duration and serum biomarker data was assessed at W0 and 4. RESULTS In total, 880 patients were enrolled (SDD/LDD = 40.6%/59.4% of patients). More SDD than LDD patients achieved absolute Psoriasis Area and Severity Index (PASI) = 0 at W28 (51.8% vs. 39.4%) and were SRes (43.7% vs. 28.1% [overall 34.4%]). SDD patients also achieved PASI = 0 quicker than LDD patients (median 141 vs. 200 days). Disease duration and prior biologic use had the greatest impact on becoming an SRe, with no strong association among these independent variables. At baseline, there were no significant differences in the serum biomarker levels of IL-17A, IL-17F, IL-22 and β-defensin 2 between SDD and LDD patients, or between SRe and non-SRe patients. Guselkumab rapidly decreased these markers of systemic inflammation across all patient groups analysed at W4. Guselkumab was well tolerated. CONCLUSIONS Guselkumab efficacy was consistent across subpopulations, on the skin and systemically. The proportion of SRes was higher in SDD than LDD patients, indicating early treatment intervention may improve clinical outcomes.
Collapse
Affiliation(s)
- K Schäkel
- Department of Dermatology, and Interdisciplinary Center for Inflammatory Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - K Reich
- Translational Research in Inflammatory Skin Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - K Asadullah
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Prof. Dr. med. Asadullah, Dermatological Practice, Potsdam, Germany
| | - A Pinter
- University Hospital Frankfurt am Main, Frankfurt, Germany
| | - D Jullien
- Department of Dermatology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
| | | | - C Paul
- Toulouse University, Toulouse, France
| | - M Gomez
- Janssen-Cilag GmbH, Neuss, Germany
| | - S Wegner
- Janssen-Cilag GmbH, Neuss, Germany
| | | | | | - Y Chen
- Janssen R&D, LLC, San Diego, USA
| | | | | | - K Eyerich
- Department of Dermatology and Venereology, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
50
|
Lian N, Chen Y, Chen S, Zhang Y, Chen H, Yang Y, Gu H, Chen Q, Li M, Chen X. Gasdermin D-mediated keratinocyte pyroptosis as a key step in psoriasis pathogenesis. Cell Death Dis 2023; 14:595. [PMID: 37673869 PMCID: PMC10482869 DOI: 10.1038/s41419-023-06094-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 08/08/2023] [Accepted: 08/21/2023] [Indexed: 09/08/2023]
Abstract
Gasdermin D (GSDMD)-mediated pyroptosis has a significant pro-inflammation characteristic due to dramatic secretion of pro-inflammatory substances. However, its role remains unclear in psoriasis as one chronic inflammatory skin disorder with high prevalence. We found that N-terminal GSDMD (N-GSDMD) was aberrantly expressed in epidermis of skin lesion in psoriasis patients and imiquimod-induced psoriasis-like dermatitis (IIPLD) mice. In epidermis of IIPLD mice and M5 (simulating psoriatic inflammatory challenge)-treated keratinocytes cultured in vitro, cleavage products of caspase-1, GSDMD and IL-1β were increased. M5-stimulated keratinocyte presented typical pyroptosis morphology accompanied with PI-staining. Gsdmd-/- keratinocytes could not present pyroptosis morphology while stimulated with M5. Electroporation of recombinant N-GSDMD could make the pyroptosis morphology reappear. In Gsdmd-/- mice or keratinocyte-specific Gsdmd conditional knockout mice, we observed the alleviation of psoriatic inflammation and epidermal aberrant expression of Ki-67 and differentiation markers (loricrin and keratin 5) after imiquimod stimulation. Transplanting skin tissue from control mice to Gsdmd-/- mice can evoke the response to imiquimod stimulation in the background of Gsdmd-/- mice (not limited in transplanting area). In M5-stimulated keratinocytes, disulfiram or GSDMD siRNA transfection can inhibit pyroptosis and eliminate disproportionate increases of Ki-67 and PI. We further validated that topically application of disulfiram (pyroptosis inhibitor) also alleviated IIPLD in mice. These findings indicate a novel mechanism that GSDMD-mediated keratinocyte pyroptosis facilitates hyperproliferation and aberrant differentiation induced by immune microenvironment in psoriatic skin inflammation, which contributes to pathogenesis of psoriasis. Our study provides an innovative insight that targeting pyroptosis can be considered as a therapeutic strategy against psoriasis.
Collapse
Affiliation(s)
- Ni Lian
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China
| | - Yujie Chen
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China
| | - Sihan Chen
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China
| | - Ying Zhang
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China
| | - Hao Chen
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China
| | - Yong Yang
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China
| | - Heng Gu
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China
| | - Qing Chen
- School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
- Department of Transfusion Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China.
| | - Min Li
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China.
- School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| | - Xu Chen
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China.
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China.
- School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| |
Collapse
|